27
c12) United States Patent Fire et al. (54) GENETIC INHIBITION BY DOUBLE-STRANDED RNA (75) Inventors: Andrew Fire, Baltimore, MD (US); Stephen Kostas, Chicago, IL (US); Mary Montgomery, St. Paul, MN (US); Lisa Timmons, Lawrence, KS (US); SiQun Xu, Ballwin, MO (US); Hiroaki Tabara, Shizuoka (JP); Samuel E. Driver, Providence, RI (US); Craig C. Mello, Shrewsbury, MA (US) (73) Assignee: Carnegie Institution of Washington, Washington, DC (US) ( *) Notice: Subject to any disclaimer, the term of this patent is extended or adjusted under 35 U.S.C. 154(b) by 1320 days. This patent is subject to a terminal dis- claimer. (21) Appl. No.: 10/283,267 (22) Filed: Oct. 30, 2002 (65) Prior Publication Data US 2003/0055020 AI Mar. 20, 2003 Related U.S. Application Data (62) Division of application No. 09/215,257, filed on Dec. 18, 1998, now Pat. No. 6,506,559. (60) Provisional application No. 60/068,562, filed on Dec. 23, 1997. (51) Int. Cl. C12N 15182 (2006.01) C12N 15190 (2006.01) Cl2N 5/10 (2006.01) (52) U.S. Cl. ....................................... 800/285; 435/468 (58) Field of Classification Search ....................... None See application file for complete search history. (56) References Cited U.S. PATENT DOCUMENTS 3,931,397 A 111976 Harnden 4,130,641 A 12/1978 Ts'o et al. 4,283,393 A 8/1981 Field 4,469,863 A 9/1984 Ts'o et al. 4,511,713 A 4/1985 Miller eta!. 4,605,394 A 8/1986 Skurkovich 4,766,072 A 8/1988 Jendrisak 4,795,744 A 111989 Carter 4,820,696 A 4/1989 Carter 4,945,082 A 7/1990 Carter 4,950,652 A 8/1990 Carter 4,963,532 A 10/1990 Carter 5,024,938 A 6/1991 Nozaki 5,034,323 A 7/1991 Jorgensen eta!. 5,063,209 A 1111991 Carter 5,091,374 A 2/1992 Carter 5,107,065 A 4/1992 Shewmaker et al. 5,132,292 A 7/1992 Carter 111111 1111111111111111111111111111111111111111111111111111111111111 AU US007622633B2 (10) Patent No.: US 7,622,633 B2 (45) Date of Patent: *Nov. 24, 2009 5,173,410 A 12/1992 Ahlquist 5,190,931 A 3/1993 Inouye 5,194,245 A 3/1993 Carter 5,208,149 A 5/1993 Inouye 5,258,369 A 1111993 Carter 5,272,065 A 12/1993 Inouye 5,365,015 A 1111994 Grierson et a!. 5,453,566 A 9/1995 Shewmaker et a!. 5,514,546 A 5/1996 Kool 5,530,190 A * 6/1996 Grierson et a!. ............. 800/283 5,578,716 A 1111996 Szyf et al. 5,593,973 A 111997 Carter 5,624,803 A 4/1997 Noonberg 5,631,148 A 5/1997 Urdea 5,643,762 A 7/1997 Ohshima et al. 5,674,683 A * 10/1997 Kool ............................. 435/6 5,683,985 A 1111997 Chu 5,683,986 A 1111997 Carter ......................... 514/44 5,691,140 A 1111997 Noren et al. 5,693,773 A 12/1997 Kandimalla 5,712,257 A 111998 Carter 5,738,985 A 4/1998 Miles 5,739,309 A 4/1998 Dattagupta 5,747,338 A 5/1998 Giese 5,795,715 A 8/1998 Livache 5,808,036 A 9/1998 Kool 5,850,026 A 12/1998 De Bonte 5,874,555 A 2/1999 Dervan 5,906,980 A 5/1999 Carter 5,908,779 A 6/1999 Carmichael 5,958,718 A 9/1999 Carter 5,972,704 A 10/1999 Draper eta!. ............... 435/375 6,010,908 A 1/2000 Gruenert et a!. 6,022,863 A 212000 Peyman ....................... 6,130,206 A 10/2000 Carter 6,133,024 A 10/2000 Helene (Continued) FOREIGN PATENT DOCUMENTS 199536778 B2 9/1995 (Continued) OTHER PUBLICATIONS 514/44 Yu et al., Plant Cell Rep., 2003, vol. 22, pp. 167-174.* (Continued) Primary Examiner-Ashwin Mehta (74) Attorney, Agent, or Firm-Morgan, Lewis & Bockius LLP (57) ABSTRACT A process is provided of introducing an RNA into a living cell to inhibit gene expression of a target gene in that cell. The process may be practiced ex vivo or in vivo. The RNA has a region with double-stranded structure. Inhibition is sequence-specific in that the nucleotide sequences of the duplex region of the RNA and of a portion of the target gene are identical. The present invention is distinguished from prior art interference in gene expression by antisense or triple- strand methods. 3 Claims, 5 Drawing Sheets

112 andrew fire - 7622633 - genetic inhibition by double-stranded rna

Embed Size (px)

DESCRIPTION

Andrew Fire, Stephen Kostas, Mary Montgomery, Lisa Timmons, SiQun Xu, Hiroaki Tabara, Samuel E. Driver, Craig C. Mello - Genetic Inhibition by Double-Stranded RNA

Citation preview

Page 1: 112   andrew fire - 7622633 - genetic inhibition by double-stranded rna

c12) United States Patent Fire et al.

(54) GENETIC INHIBITION BY DOUBLE-STRANDED RNA

(75) Inventors: Andrew Fire, Baltimore, MD (US); Stephen Kostas, Chicago, IL (US); Mary Montgomery, St. Paul, MN (US); Lisa Timmons, Lawrence, KS (US); SiQun Xu, Ballwin, MO (US); Hiroaki Tabara, Shizuoka (JP); Samuel E. Driver, Providence, RI (US); Craig C. Mello, Shrewsbury, MA (US)

(73) Assignee: Carnegie Institution of Washington, Washington, DC (US)

( *) Notice: Subject to any disclaimer, the term of this patent is extended or adjusted under 35 U.S.C. 154(b) by 1320 days.

This patent is subject to a terminal dis­claimer.

(21) Appl. No.: 10/283,267

(22) Filed: Oct. 30, 2002

(65) Prior Publication Data

US 2003/0055020 AI Mar. 20, 2003

Related U.S. Application Data

(62) Division of application No. 09/215,257, filed on Dec. 18, 1998, now Pat. No. 6,506,559.

(60) Provisional application No. 60/068,562, filed on Dec. 23, 1997.

(51) Int. Cl. C12N 15182 (2006.01) C12N 15190 (2006.01) Cl2N 5/10 (2006.01)

(52) U.S. Cl. ....................................... 800/285; 435/468 (58) Field of Classification Search ....................... None

See application file for complete search history.

(56) References Cited

U.S. PATENT DOCUMENTS

3,931,397 A 111976 Harnden 4,130,641 A 12/1978 Ts'o et al. 4,283,393 A 8/1981 Field 4,469,863 A 9/1984 Ts'o et al. 4,511,713 A 4/1985 Miller eta!. 4,605,394 A 8/1986 Skurkovich 4,766,072 A 8/1988 Jendrisak 4,795,744 A 111989 Carter 4,820,696 A 4/1989 Carter 4,945,082 A 7/1990 Carter 4,950,652 A 8/1990 Carter 4,963,532 A 10/1990 Carter 5,024,938 A 6/1991 Nozaki 5,034,323 A 7/1991 Jorgensen eta!. 5,063,209 A 1111991 Carter 5,091,374 A 2/1992 Carter 5,107,065 A 4/1992 Shewmaker et al. 5,132,292 A 7/1992 Carter

111111 1111111111111111111111111111111111111111111111111111111111111

AU

US007622633B2

(10) Patent No.: US 7,622,633 B2 (45) Date of Patent: *Nov. 24, 2009

5,173,410 A 12/1992 Ahlquist 5,190,931 A 3/1993 Inouye 5,194,245 A 3/1993 Carter 5,208,149 A 5/1993 Inouye 5,258,369 A 1111993 Carter 5,272,065 A 12/1993 Inouye 5,365,015 A 1111994 Grierson et a!. 5,453,566 A 9/1995 Shewmaker et a!. 5,514,546 A 5/1996 Kool 5,530,190 A * 6/1996 Grierson et a!. ............. 800/283 5,578,716 A 1111996 Szyf et al. 5,593,973 A 111997 Carter 5,624,803 A 4/1997 Noonberg 5,631,148 A 5/1997 Urdea 5,643,762 A 7/1997 Ohshima et al. 5,674,683 A * 10/1997 Kool ............................. 435/6 5,683,985 A 1111997 Chu 5,683,986 A 1111997 Carter ......................... 514/44 5,691,140 A 1111997 Noren et al. 5,693,773 A 12/1997 Kandimalla 5,712,257 A 111998 Carter 5,738,985 A 4/1998 Miles 5,739,309 A 4/1998 Dattagupta 5,747,338 A 5/1998 Giese 5,795,715 A 8/1998 Livache 5,808,036 A 9/1998 Kool 5,850,026 A 12/1998 De Bonte 5,874,555 A 2/1999 Dervan 5,906,980 A 5/1999 Carter 5,908,779 A 6/1999 Carmichael 5,958,718 A 9/1999 Carter 5,972,704 A 10/1999 Draper eta!. ............... 435/375 6,010,908 A 1/2000 Gruenert et a!. 6,022,863 A 212000 Peyman ....................... 6,130,206 A 10/2000 Carter 6,133,024 A 10/2000 Helene

(Continued)

FOREIGN PATENT DOCUMENTS

199536778 B2 9/1995

(Continued)

OTHER PUBLICATIONS

514/44

Yu et al., Plant Cell Rep., 2003, vol. 22, pp. 167-174.*

(Continued)

Primary Examiner-Ashwin Mehta (74) Attorney, Agent, or Firm-Morgan, Lewis & Bockius LLP

(57) ABSTRACT

A process is provided of introducing an RNA into a living cell to inhibit gene expression of a target gene in that cell. The process may be practiced ex vivo or in vivo. The RNA has a region with double-stranded structure. Inhibition is sequence-specific in that the nucleotide sequences of the duplex region of the RNA and of a portion of the target gene are identical. The present invention is distinguished from prior art interference in gene expression by antisense or triple­strand methods.

3 Claims, 5 Drawing Sheets

Page 2: 112   andrew fire - 7622633 - genetic inhibition by double-stranded rna

US 7,622,633 B2 Page 2

U.S. PATENT DOCUMENTS

6,136,601 A 6,291,504 B1 6,369,038 B1 6,372,965 B1 6,506,559 B1 6,531,647 B1 6,573,099 B2 6,635,805 B1

10/2000 Meyer, Jr. et a!. 9/2001 Nugiel 4/2002 Blumenfeld 4/2002 Lightner 112003 Fire 3/2003 Baulcombe 6/2003 Graham ...................... 435/455

10/2003 Baulcombe 2003/0061626 A1 2004/0138168 A1

3/2003 Plaetinck et al ................ 800/8 7/2004 Satishchandran

AU AU CA CA CN EP EP EP EP EP EP EP EP EP EP EP EP EP EP GB GB GB GB JP wo wo wo wo wo wo wo wo wo wo wo wo wo wo wo wo wo wo wo wo wo wo wo wo wo wo wo wo

FOREIGN PATENT DOCUMENTS

199929163 B2 200195225

2012312 2370628

00806369 0318281 A2 0325018 A2 0347501 A1 0213921 B1 0495831 B1 0286224 B1 0350151 B1 0303516 B1 0306347 B1 0465572 B1 0281380 0308066 B1 0300680 B1

983370 B1 9710475.6

2353282 2362885 A 2377221 B

09-227413 wo 90/14090 wo 90/12094 wo 90/12488 wo 91105559 wo 91118611 wo 92/18522 wo 93/23569 wo 94/01550 wo 95/03406 wo 95/15378 wo 95/18223 wo 95/18854 wo 95/34668 wo 96/35706 wo 97/10360 wo 97/11170 wo 97/44450 wo 98/05770 wo 98/53083 wo 98/53083 wo 99/32619 wo 99/49029 wo 99/53050 wo 99/61631 wo 00/01846 wo 00/63364

wo 011070949 wo 04/022748

3/1999 1112001 3/1990 4/2000 4/2000 5/1989 7/1989

12/1989 8/1990 9/1990

1111992 3/1994 7/1994 5/1995 6/1995

1111995 12/1995 9/1996 9/2003 7/1997 2/2001

12/2001 8/2004 9/1997 5/1989

10/1990 1111990 5/1991

12/1991 10/1992 1111993

111994 2/1995 6/1995 7/1995 7/1995

12/1995 1111996 3/1997 3/1997

1111997 2/1998 5/1998

1111998 7/1999 9/1999

10/1999 12/1999

1/2000 10/2000 9/2001 3/2004

OTHER PUBLICATIONS

Horiguchi, G., Different., 2004, vol. 72, pp. 65-73.* Kumagai et a!., Proc. Nat!. Acad. Sci., USA, 1995, vol. 92, pp. 1679-1683.* Holtorf et al., Plant Mol. Bioi., 1995, vol. 29, pp. 637-646.*

Klein eta!. (Nature, 1987, vol. 327, pp. 70-73.* Brisson eta!., Nature, 1984, vol. 310, pp. 511-514.* McCabe eta!., Biotech., 1988, vol. 6, pp. 923-926. * Metzlaff eta!., Cell, Mar. 21, 1997, vol. 88, pp. 845-854.* van der Meer eta!., Plant Mol. Bioi., 1990, vol. 15, pp. 95-109.* Grayburn eta!., Plant Cell Rep., 1995, vol. 14, pp. 285-289.* Cameron eta!. ( 1991 ), "Inhibition of gene expression by a short sense fragment," Nucl Acids Res 19(3):469-475. Agrawal, "Antisense Oligonucleotides: Towards Clinical Trials" (1996), Tibtech 14: 376-387. Anderson, "Human Gene Therapy" (1998), Nature, 392: 25-30. Branch, "A Good Antisense Molecule Is Hard to Find" (1998), TIES 23: 45-50. Clemens et a!., Use of Double-Stranded RNA Interference in Drosophila Cell Lines to Dissect Signal Transduction Pathways (1999), PNAS 97: 6499-6503. Mercola and Cohen, "Antisense Approaches to Cancer Gene Therapy" (1995), Cancer Gene Therapy 2: 47-59. Sharp, "RNAi and Double-Strand Rna" (1999), Genes & Develop­ment 13: 139-141. Verna and Somia, "Gene Therapy-Promises, Problems and Pros­pects" (1997), Nature 389: 239-242. Bevec eta!., "Constitutive expression of chimeric neo-Rev response element transcripts suppresses HIV-1 replication in human CD4+ T lymphocytes" (1994) Hum Gene Ther 5:193-201. Cohli et al., "Inhibition of HIV-1 multiplication in a human CD4+ lymphocytic cell line expressing antisense and sense RNA molecules containing HIV-1 packaging signal and Rev response element(s)" (1994) Antisense Res Dev. 4:19-26. Dorer eta!., "Expansions oftransgene repeats cause heterochromatin formation and gene silencing in Drosophila" (1994) Cell 77:993-1002. Dorer et a!., "Transgene repeat arrays interact with distant heterochromatin and cause silencing in cis and trans" (1997) Genet­ics 147:1181-1190. Fraser eta!., "Effects of c-myc first exons and 5' synthetic hairpins on RNA translation in oocytes and early embryos of Xenopus laevis"(1996) Oncogene 12:1223-1230. Good et a!., "Expression of small, therapeutic RNAs in human cell nuclei" (1997) Gene Therapy 4:45-54. Kozak "Circumstances and mechanisms of inhibition of translation by secondary structure in eucaryotic mRNAs" (1989) Mol Cell Bioi 9:5134-5142. Liebhaber eta!., "Translation inhibition by an mRNA coding region secondary structure is determined by its proximity to the AUG ini­tiation codon" (1992) J Mol Bio/226:609-621. Lingelbach & Dobberstein "An extended RNA/RNA duplex struc­ture within the coding region of mRNA does not block translational elongation" (1998) Nucleic Acids Res 16:3405-3414. Lisziewicz eta!., "Inhibition of human immunodeficiency virus type 1 replication by regulated expression of a polymeric Tat activation response RNA decoy as a strategy for gene therapy in AIDS" (1993) Proc Nat! A cad Sci 90: 8000-8004. Loomis et al., "Antisense RNA inhibition of expression of a pair of tandemly repeated genes results in a delay in cell-cell adhesion in Dictyostelium" ( 1991) Antisense Research and Development 1:255-260. Mikoshiba eta!., "Molecular biology of myelin basic protein: gene rearrangement and expression of anti -sense RNA in myelin-deficient mutants" (1991) Comp Biochem Physiol1:51-61. Okano eta!., "Myelin basic protein gene and the function of antisense RNA in its repression in myelin-deficient mutant mouse" (1991) Journal of Neurochemistry 56:560-567. Pelletier et a!., "Insertion mutagenesis to increase secondary struc­ture within the 5' noncoding region of a eukaryotic mRNA reduces translational efficiency"(1985) Cell40:515-526. Sijen et a!., "RNA-Mediated Virus Resistance: Role of Repeated Transgenes and Delineation of Targeted Regions" (1996) Plant Cell 8:2277-2294. Sun et al., "Resistance to human immunodeficiency virus type 1 infection conferred by transduction of human peripheral blood lym-

Page 3: 112   andrew fire - 7622633 - genetic inhibition by double-stranded rna

US 7,622,633 B2 Page 3

phocytes with ribozyme, antisense, or polymeric transactivation response element constructs" (1995) Proc Nat! Acad Sci 92:7272-7276. Watson, "A new revision of the sequence of plasmid pBR322" ( 1988) Gene 70:399-403. Weaver et a!., "Introduction by molecular cloning of artifactual inverted sequences at the 5' terminus of the sense strand of bovine parathyroid hormone eDNA" (1981) Proc Nat! Acad Sci 78:4073-4077. Agrawal, Sundhir (1996) "Antisense Oligonucleotides: Towards Clinical Trials" Trends in Biotechnology 14, p. 376. Akgun, Ercan ( 1997) "Palindrome Resolution and Recombination in the Manunalian Germ Line "Molecular and Cellular Biology 17(9): 5559. Akhtar, S. ( 1996) "Anti-HIV therapy with antisense oligonucleotides and ribozymes realistic approaches or expensive myths?" Journal of Antimicrobial Chemotherapy p. 159. Awram P, Gardner RC, Forster RL, Bellamy AR. "The potential of plant viral vectors and transgenic plants for subunit vaccine produc­tion" AdvVirus Res. 2002;58:81-124. Bahner, Ingrid (1996) "Transduction of Human CD34+ Hematopoietic Progenitor Cells by a Retroviral Vector Expressing an RRE Decoy Inhibits Human Immunodeficiency Type 1 Replication in Myelomonocytic Cells Produced in Long-Term Culture" Journal ofVirology 70(7): 4352. Balmori-Melian E, MacDiarmidRM, Beck DL, Gardner RC, Forster RL. "Sequence-, tissue-, and delivery-specific targeting of RNA dur­ing post-transcriptional gene silencing" Mol Plant Microbe Interact. Aug. 2002;15(8):753-63. Barbeau, Benoit (1996) "Characterization of the Human and Mouse Fli-1 promoter regions" BBA Gene Structure and Expression 1307 p. 220. Baulcombe DC (1996) "Mechanisms of Pathogen-Derived Resis­tance to Viruses in Transgenic Plants" Plant CellS: 1833-1844. Beck DL, Forster RL, Bevan MW, Boxen KA, Lowe SC. "Infectious transcripts and nucleotide sequence of cloned eDNA of the potexvirus white clover mosmc virus" Virology. Jul. 1990; 177( 1 ): 152-8. Beck DL, Guilford PJ, Voot DM, Andersen MT, Forster RL. "Triple gene block proteins of white clover mosaic potexvirus are required for transport" Virology. Aug. 1991;183(2):695-702. Beck DL, Van Dolleweerd CJ, Lough TI, Balmori E, Voot DM, Andersen MT, O'Brien IE, Forster RL. "Disruption of virus move­ment confers broad -spectrum resistance against systemic infection by plant viruses with a triple gene block" Proc Nat! Acad Sci US A. Oct. 25, 1994;91(22):10310-10314. Betz, Natalie (2003) "RNAi: RNA Interference" RNAi: RNA Inter­ference 83: 33. Bigler, Jeanette (1995) "Novel Location and Function of a Thyroid Hormone Response Element" The EMBO Journal14(22): 5710. Bingham, Paul (1997) "Cosupression Comes to the Animals" Cell 90: 385. Bisat, F.N.B.K. Raj and P.M. Pitha ( 1998) "Differential and Cell Type Expression of Murine Alpha-Interferon Genes is Regulated on the Transcriptional Level" Nucleic Acids Research 16(13): 6067. Brown, David (1993) "Identification tluough Overexpression and Tagging of the Variant Type of the Mouse H1e and H1c Genes" The Journal of Biological Chemistry 268(1): 713. Bryan GT, Gardner RC, Forster RL. (1992) "Nucleotide sequence of the coat protein gene of a strain of clover yellow vein virus from New Zealand: conservation of a stem-loop structure in the 3' region of potyviruses" Arch Virol124(1-2):133-46. Buchan et al. (1994) "Characterization of three non-peptide endothelin receptor ligands using human cloned ETA and ETB recep­tors" Br. J. Pharmacol. 112: 1251-1257. Chernajovsky, Yuti (1996) "Human Kinesin Light (b) Chain Gene: DNA Sequence and Functional Characterization oflts Promoter and First Exon" DNA and Cell Biology 15: 965. Christy, Robert (1988) "Functional Analysis of the Long Terminal Repeats of Intracisternal A-Particle Genes: Sequences withing the U3 Region Determine Both the Efficiency and Direction of Promoter Activity" Molecular and Cellular Biology 8(3): 1093.

Clusel, Catherine (1993) "Ex Vivo Regulation of Specific Gene Expression by Nanomolar Concentration of Double-Stranded Dumbell Oligonucleotides" Nucleic Acids Research 21(15): 3405. Clusel, Catherine (1995) "Inhibition of HSV-1 Proliferation by Decoy Phosphodiester Oligonucleotides Containing ICP4 Recogni­tion Sequences" Gene Expression 4: 301. DeCoy, Donald (1995) "Anti sense DNA Down-regulates Protein Kinase C and Enhances Vasopression-stimulated Na Absorption in Rabbit Cortical Collecting Duct" J. Clin. Invest. 95: 2749. Dobrikova eta!. (1996) "T7 DNA-dependent RNA polymerase can transcribe RNA from tick-borne encephalitis virus (TBEV) eDNA with SP6 promoter" FEBS Letters 382, pp. 327-329. Dougherty WG and Parks TD (1995)"Transgenes and gene supres­sion: telling us something new?'' Curr Opin Cell Bioi. 7(3):399-405. Driver SE, Ali M, Mello CC (1996) "Formation of Heritable Anti­Transgenes After RNA Microinjection inC elegans" 1996 East Coast Worm Meeting abstract 33. Driver SE, Ali, M, Mello CC (1997) ""Antisense" In C. Elegans Heritable Gene Silencing Induced By RNA Microinjection" Early 1997 International Worm Meeting abstract 136. Dronkert, Mies (2000) "Mouse RAD54 Affects DNA Double-Strand Break Repair and Sister Chromatid Exchange" Molecular and Cel­lular Biology 20(9): 3147. Dykxhoorn DM eta!., (2003) "Killing the messenger: Short RNAs that silence gene expression" Nat Rev Mol Cell Bioi. Jun.;4(6):457-67. Eagles RM, Gardner RC, Forster RL "Nucleotide sequence of the tamarillo mosaic virus coat protein gene" Nucleic Acids Res. Dec. 11, 1990; 18(23): 7166. Eagles, R. M., E. Balmori-Melian, D. L. Beck, R. C. Gardner, and R. L. Forster. 1994. "Characterization of NTPase, RNA-binding and RNA-helicase activities of the cytoplasmic inclusion protein of tamarillo mosaic potyvirus" Eur. J. Biochem. 224:677-684. Elroy-Stein eta!. (1990) "Cytoplasmic expression system based on constitutive synthesis ofbacteriophage T7 RNA polymerase in mam­malian cells" Pro. Nat!. Acad. Sci. USA 87, pp. 6743-6747. Escude, Christophe (1996) "Stable triples helics formed by oligonucleotide N3'-P5' phosphoramidates inhibit transcription elongation" Proc. Nat!. Acad. Sci. USA 93: 4356. Everett KR, Milne KS, Forster RL. "Nucleotide sequence of the coat protein genes of strawberry latent ring spot virus: lack ofhomologyto the nepoviruses and comoviruses" J Gen Virol. Jul. 1994;75 ( Pt 7): 1821-5. Faruqi, Tatjana R. (1997) "IFN-y Inhibits Double-Stranded RNA­Induced E-Selectin Expression in Human Endothelial Cells" The Journal oflmmunology 159(8): 3989. Fiaschi, Tania (1997) "The 5'-untranslated Region of the Human Muscle Acylphosphatase m RNA has an Inhibitory Effect on Protein Expression" FEBS Letters 417 No. 1,3 p. 130. Finkler, Aliza ( 1992) "Immunity and Resistance to the KP6 Toxin of Ustilago Maydis" MGG 233(3): 395. Forster RL, Beck DL, Guilford PJ, Voot DM, Van Dolleweerd CJ, Andersen MT. ( 1992) "The coat protein of white clover mosaic potexvirus has a role in facilitating cell-to-cell transport in plants" Virology 191(1):480-4. Forster RL, Bevan MW, Harbison SA, Gardner RC. (1988) "The complete nucleotide sequence of the potexvirus white clover mosaic virus" Nucleic Acids Res. 16( 1 ):291-303. Foster TM, Lough T J, Emerson SJ, Lee RH, Bowman JL, Forster RL, Lucas WJ. (2002) "A surveillance system regulates selective entry of RNA into the shoot apex" Plant Cell. 14(7):1497-508. Fuerst eta!. ( 1986) "Eukaryotic transient-expression system based on recombinant vaccinia virus that synthesizes bacteriophage T7 RNA polymerase" Pro. Nat!. Acad. Sci. USA 83: 8122-8126. Gao eta!. (1997) "Human genes encoding U3 snRNA associate with coiled bodies in interphase cells and are clustered on chromosome 17p11.2 in a complex inverted repeat structure" Nucleic Acids Research 25(23): 4740-4747. Gardiner SE, Bassett HCM, Noiton DAM, Bus VG, Hofstee ME,White AG, Ball RD, Forster RLS, Rikkerink EHA (1996) "Adetailed linkage map around an apple scab resistance genedemonstrates that two disease resistance classes carry the Vfgene" Theor Appl Genet 93:485-493.

Page 4: 112   andrew fire - 7622633 - genetic inhibition by double-stranded rna

US 7,622,633 B2 Page 4

Garrick, David 1998 "Reseat-induced gene silencing in mammals" Nature Genetics 18: 56. Gervaix, Alain (1997) "Multigene Antiviral Vectors Inhibit Diverse HIV Virus Type 1 Clades" Journal ofVirology 71(4): 3048. Gessani, Sandra ( 1989) "Activators of Protein Kinase C Enhance Accumulation oflnterferon-B mRNA in Murine Cell Lines" Jour­nal oflnterferon Research 9: 543. Gimmi, Edward R. ( 1989) "Alterations in the pre-mRNA topology of the bovine growth hormone polyadenylation region decrease poly (A) site efficiency" Nucleic Acids Research 17(17): 6983. Giovannangeli, Carine ( 1996) "Accessibility of nuclear DNA triplex­forming oligonucleotides: The integrated HIV-1 provirus as a target" Proc. Nat!. Acad. Sci., USA 94: 79. Graham, Geoffrey (1990) "RNA Transcripts of the HIV Transactiva­tion Response Element Can Inhibit Action of the Viral Transactiva­tor" Proc. Nat!. Acad. Sci. USA 87: 5817. Graham, Geoffrey (1992) "A Rapid and Reliable Method to Create Tandem Arrays of Short DNA Sequences" Bio Techniques (Research Report) 13(5): 780. Groger, Richard ( 1989) "Directional antisense and sense eDNA clon­ing using Epstein-Barr virus episomal expression vectors" Gene (81 ): 285. Guilford PJ, Beck DL, Forster RL. (1991) "Influence of the poly( A) tail and putative polyadenylation signal on the infectivity of white clover mosaic potexvirus"Virology 182(1):61-7. Hacker eta!. (1995) "Expression of Sry, the mouse sex determining gene" Development 121, pp. 1603-1614. Haines, Dale ( 1991) "Cellular Response to Double-Stranded RNA" Journal of Cellular Biochemistry46 1991 p. 9. Harbison SA, Forster RL, Guilford PJ, Gardner RC. (1988) "Orga­nization and interviral homologies of the coat protein gene of white clover mosaic virus" Virology 162(2):459-65. Harcourt, Brian ( 1998) "Ebola Virus Inhibits Induction of Genes by Double-Stranded RNA in Endothelial Cells" Virology 252 (article No. VY989446) p. 179. Helliwell CA, Olive MR, Gebbie L, Forster R, Peacock WJ, Dennis ES (2000) "Isolation of an ent-kaurene oxidase eDNA from Cucurbita maxima" Aust J Plant Physiol27: 1141-1149. Henderson, Samuel (1993) "Instability of a Plasmid-Borne Inverted Repeat in Saccharomyces cerevisiae" Genetics 133 : 57. Hirashima, Akikazu (1986) "Engineering of the mRNA-interfering complementary RNA immune system against viral infection" Proc. Nat!. Acad. Sci. USA 83: 7726. Sawaki, Hirashima (1989) "Artificial immune system against infec­tion involving antisense RNA targeted to the 5'-terminal noncoding region of coliphage RNA" Journal of Biochemistry 106(1): 163. Howitt RL, Beever RE, Pearson MN, Forster RL. "Genome charac­terization of Botrytis virus F, a flexuous rod-shaped mycovirus resembling plant 'potex-like' viruses" J Gen Virol. Jan. 2001;82(Pt 1):67-78. Imazeki eta!. ( 1988) "Integrated structures of duck hepatitis B virus DNA in hepatocellular carcinoma" Journal of Virology 62(3) 861-865. Krystal, Geoffrey ( 1988) "Multiple Mechanisms for Transcriptional Regulation of the myc Gene Family in Small-Cell Lung Cancer" Molecular and Cellular Biology 8(8): 3373. Krystal, Geoffrey (1990) "N-myc mRNA Forms an RNA-RNA Duplex with Endogenous Antisense Transcripts" Molecular and Cel­lular Biology 10(8): 4180. Lee et a!. ( 1993) "The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14" Cell 75: 843-854. Lee, Seok-Woo (1994) "Inhibition ofHIV Type 1 in Human T Cells by a potent rev response element decoy consisting of the 13-Nucleotide Minimal Rev-Binding Domain" Journal of Virology 68(12): 8254. Lee, Seok-Woo ( 1996) "The Hemagglutinin Genes hagB and hagC of Porphyromonas gingiva/is Are Transcribed in Vivo as Shown by Use of a New Expression Vector" Infection and Immunity 64 (11): 4802. Liefting LW, Andersen MT, Beever RE, Gardner RC, Forster RL. (1996) "Sequence heterogeneity in the two 16S rRNA genes of Phormium yellow leaf phytoplasma" Appl Environ Microbiol. 62(9):3133-9.

Lindbo eta!. ( 1993) "Induction of a Highly Specific Antiviral State in Transgenic Plants: Implications for Regulation of Gene Expression and Virus Resistance" the Plant Cell vol. 5, Issue 12 1749-1759. Lough TJ, Balmori E, Beck DL, Forster RL. "Western analysis of transgenic plants" Methods Mol Bioi. 1998;81:447- 51. Lough TJ, Emerson SJ, Lucas WJ, Forster RL. "Trans-complementa­tion of long-distance movement ofWhite clover mosaic virus triple gene block (TGB) mutants: phloem-associated movement of TGBpl"Virology. Sep. 15, 2001;288(1):18-28. Lough TJ, Netzler NE, Emerson SJ, Sutherland P. Carr F, Beck DL, Lucas WJ, Forster RL. "Cell-to-cell movement ofpotexviruses: evi­dence for a ribonucleoprotein complex involving the coat protein and first triple gene block protein" Mol Plant Microbe Interact. Sep. 2000; 13(9):962-74. Lough, T.J., Shash, K., Xoconostle-Cazares, B., Hofstra, K.R., Beck, D.L., Balmori, E., Forster, R.L.S., and Lucas, W.J. (1998). "Molecu­lar dissection of the mechanism by which potexvirus triple gene block proteins mediate cell-to-cell transport of infectious RNA" Mol. Plant-Microbe Interact. 11: 801-814. Mace eta!. (1991) "Interferon-regulated viral replication in chroni­cally HIV !-infected promonocytic U937 cells" Research in Virology 142: 213-220. Matthieu (1992) "Myelin-Deficient Mutant Mice an in Vivo Model for Inhibition of Gene Expression by Natural Antisense RNA" Annals of the New York Academy of Sciences 660: 188. Mayne, Lynne ( 1998) "SV 40-transformmednormal and DNA-repair deficient human fibroblasts can be transfected with high frequency but retain only amounts of integrated DNA" Gene 66: 65. McCormack, Stephen (1992) "Mechanism of Interferon Action: Identification of a RNA Binding Domain within the N-terminal Region of the Human RNA-Dependent PllelF-2a Protein Kinase" Virology 188: 47. McNair, A.N. B. (1994) "Hepatitis delta virus replication in vitro is not affected by interferon-x or-y despite intact cellular responses to interferon and dsRNA" Journal of General Virology 75: 1371. Mikoshiba, Katshuhiko (1990) "Chimeric and Molecular Genetic Analysis of Myelin-Deficient (Shiverer and Mid) Mutant Mice" Annals of the New York Academy of Sciences 605: 166. Mikoshiba, Katshuhiko (1990) "Molecular Biology of Myelin Basic Protein Gene Rearrangement and Expression of Anti-Sense RNA in Myelin-Deficient Mutants" Comparative Biochemistry and Physiol­ogy 98C( 1 ): 51. Moore CJ, Sutherland PW, Forster RL, Gardner RC, MacDiarmid RM. "Dark green islands in plant virus infection are the result of posttranscriptional gene silencing" Mol Plant Microbe Interact. Aug. 14, 2001; (8):939-46. Morishita, Ryuichi (1996) "Role of Transcriptional cis-Elements, Angiotensinogen Gene Activating Elements, of Angiotensinogen Gene Blood Pressure Regulation" Hypertension 27, 1996 p. 502. Morris, Kevin (2004) "Small Interfering RNA-Induced Transcrip­tional Gene Silencing in Human Cells" Science 305: 1289. Nagy, Eszter (1995) "Glyceraldehyde 3-phosphate Dehydrogenase Selectively Binds AU-rich RNA in the NAD +binding Region (Rossmann Fold)*" the Journal of Biochemical Chemistry 270(6): 2755. Noguchi, Masayuki (1994) "Characterization of an Antisense Inr Element in the eiF-2a Gene" The Journal of Biological Chemistry 269(46): 29161. Pal-Bhadra, Manika (1997) "Cosupression in Drosophila: Gene Silencing of Alcohol dehydrogenes by white-Adh Transgenes Is Polycomb Dependent" Cell90, Aug. 1997 p. 479. Palmiter eta!. (1984) "Transmission distortion and mosaicism in an unusual transgenic mouse pedigree" Cell36 pp. 869-877. Pe'ery, Tsafrira (1997) "Synthesis and Purification of Single­Stranded RNA for use in Experiments with PKR and in Cell-Free Translation Systems" Methods: A Companion to Methods in Enzy­mology 11, p. 371. Peyman, John ( 1997) "Molecular Biology and the Vascular Surgeon" Basic Science of Vascular Disease (Chapter 17): 17. Raponi, Mitch (2003) "Double-stranded RNA-mediated gene silenc­ing in Fission yeast" Nucleic Acids Research 31( 15), p. 448.

Page 5: 112   andrew fire - 7622633 - genetic inhibition by double-stranded rna

US 7,622,633 B2 Page 5

Resnekov, Orna ( 1989) "RNA Secondary Structure is an Integral Part of the in Vitro Mechanism of Attenuation in Simian Virus 40" The Journal of Biological Chemistry 264( 17): 9953. Reuben et a!. ( 1994) "Cloning and expression of the rabbit gastric CCK-A receptor" Biochimica et Biophysica Actya, 1219: 3 21-3 27. Rocheleau eta!. (1997) "Wnt signaling and an APC-related gene specify endoderm in early C. elegans embryos" Cell90(4):707-716. Rodriguez eta!. ( 1990) "Regulated expression of nuclear genes byT3 RNA polymerase and lac repressor, using recombinant vaccinia virus vectors" Journal ofVirology 64( 1 0): 4851-4857. Roy, Patrice (1990) "Effect of mRNA secondary structure on the efficiency of translational initiation by eukaryotic ribosomes" Euro­pean Journal of Biochemistry 191(3): 647. Ruskin, Barbara (1993) "Mutations in POLl Increase the Mitotic Instability ofTandem Inverted Repeats in Saccharomyces cerevisiae, "Genetics 133: 43. Sabl, Joy ( 1996) "Copy Number and Orientation Determine the Sus­ceptibility of a Gene to Silencing by Nearby Hetreochrornatin in Drosophila" Genetics 142, Feb. 1996 p. 447. Schmitt, Hans Peter (1986) "Characterization of cloned sequences complementary to F9 cell double-stranded RNA and their expression during differentiation" Differentiation 30: 205. Shaffer, Catherine (2004) "RNAi Shakes up Bio BEO Investor Con­ference" Biotech News 24(8): 30. Silverman eta!. (1992) "Role of sequences within the first intron in the regulation of expression of eukaryotic initiation factor 2 alpha" The Journal of Biological Chemistry 267(14): 9738-9742. Simons, Robert (1988) "Naturally occurring antisense RNA control-a brief review" Gene 72: 35. Smith, Neil (2000) "Total Silencing by inton-spliced hairpin RNAs" Nature 407: 319. Smolinski, Paula (1995) "Double-Stranded RNA Induces Sickel Erythrocyte Adherence to Endothelium: A Potential Role for Viral Infection in Vaso-Occlusive Pain Episodes in Sickel Cell Anemia" Blood 85(10): 2945. Smythe, Jason (1995) "Gene therapeutic agents: The use of ribozymes, antisense, and RNA decoys for HIV-1 infection" Inflanun Res 44: 11. Sonoda, Ken go ( 1996) "Asymmetric deletion of the junction between the short unique region and the inverted repeat does not affect viral growth in culture and vaccine-induced immunity against Marek's disease" Vaccinee 14(4): 277. Stevenson M (2003) "Dissecting HIV-1 through RNA interference" Nat Rev Immunol. 3(11):851-8. Sullenger, Bruce (1990) "Overexpression of TAR Sequence Renders Cells Resistant to HIV Replication" Cell 63: 601. Sullenger, Bruce (1991) "Analysis oftrans-Acting Response Decoy RNA-Meditated Inhibition of HIV Type 1 Transactivation" Journal ofVirology vol. 65( 12): 6811. Sun, Lun-Quan (1994) "Ribozyme-meditated suppression of Moloney murine leukemia virus and HIV tyoe I replication in per­missive cell lines" Proc. Nat!. Acad. Sci USA 91: 9715. Symington, Lorraine (2002) "Role ofRAD52 Epistasis Group Genes in Homologous Recombination and Double-Strand Break Repair" Microbiology and Molecular Biology Reviews 66(4): 630. Tanaka eta!. (1994) "Sequence-specific interaction of alpha-beta­anomeric double-stranded DNA with the p50 subunit ofNF kappa B: application to the decoy approach" Nucleic Acids Research 22(15): 3069-3074. Taylor CB (1997) "Comprehending Cosuppression" Plant Cell 9: 1245-1249. Tosic, Mirjana (1990) "Post-transcriptional events are responsible for Joe expression of myelin basic protein in myelin deficient mice: role of natural antisense RNA" The EMBO Journal9(2): 401. U sdin eta!. ( 1993) "SP6 RNA polymerase containing vaccinia virus for rapid expression of cloned genes in tissue culture" BioTechniques 14(2): 222-224. Van Steeg, Harry (1991) "The translation in vitro of rat ornithine decarboxylase m RNA is blocked by its 5" untranslated region in a polyamine-independent way" The Biochemical Journal 274: 274.

Volloch, Vladimir Z. ( 1994) "Evolutionary conserved elements in the 5' untranslatedregions ofB globin mRNAmediate site-specific prim­ing of a unique hairpin structure during eDNA synthesis" Nucleic Acids Rsearch 22(24): 5302. Wang et a!. (1994) "An unusual nucleoporin-related messenger ribonucleic acid is present in the germ cells of rat testis" Biology of Reproduction 51: 1022-1030. Wang, Lili (1997) "A factor IX-deficient mouse model for hemp­philia B gene therapy" Proc. Nat!. Acad. Sci USA,vol. 94: 11563. Webster DE, Guy PL, Beck DL, Forster RL. ( 1999) "Distribution and diversity of New Zealand isolates of ryegrass mosaic virus" Arch Virol. 144(10):2059-2064. Williams, Trevor ( 1986) "A mouse locus at which transcription from both DNA strands produces mRNAs complementary at their 3' ends" Nature 322: 275. Wolffe, Alan P. ( 1997) "Transcription Control: Repressed repeats express themselves" Current Biology 7(12): R796. Wu, Chaoquin ( 1994) "Interferon-Stimulated Response Element and NFkB Sites Cooperate to Regulate Double-Stranded RNA-Induced Transcription of the IP-10 Gene" Journal oflnterferon Research 14: 357. Wu, Shiyong (1996) "Double stranded (ds) RNA binding and not Dimerization Correlates with the Activation of the dsRNA-depen­dent Protein Kinase (PKR)" The Journal of Biochemical Chemistry 271(3): 1756. Xiong et al. (1995) "Signaling properties of mouse and human corticotroph-releasing factor (CRF) receptors: decreased coupling efficiency of human type II CRF receptor" Endocrionlogy 136(5): 1828-1834. Yarney eta!. (1993) "Molecular cloning and expression of the ovine testicular follicle stimulating hormone receptor" Molecular and Cel­lular Endocrinology 93: 219-226. Yu, Mang ( 1994) "Progress towards gene therapy for HIV infection" Gene Therapy 1: 13. Zakharyan, R.A. (1986) "Stimulation of Double-Spiral RNA Trans­formation of Prokaryotic and Eukaryotic Cells" Doklady Akadem: Nauk SRR 288(5): 1251. Zhenhua, Jin (1991) "Expression of Firefly Luciferase Gene in Xenopus laevis oocyte" Chinese Journal ofBiotechnology 7(4): 279-284. Marcus eta!., Pfomega Notes Magazine, No. 58, 1996. p. 36. Agrawal, Sudhir eta!. (1995) "Self-Stabilized Oligonucleotides as Novel Antisense Agents" pp. 105-120. In: Akhtar S, ed. Delivery strategies: antisense oligonucleotide therapeutics. Boca Raton, FL: CRC Press. Angell, S.M., eta!. (1997) "Consistent gene silencing in transgenic plants expressing a replicating potato virus X RNA", EMBO Journal, 16(12), pp. 3675-3684. Baum, E. Z., et a!. (1983) "Inhibition of protein synthesis in reticulocyte lysates by a double-stranded RNA component in HeLa mRNA" Biochem. Biophys. Res. Commun. 114(1), pp. 41-49. Bhan, P., et al. (1997) "2',5'-Linked oligo-3'-deoxyribonucleoside phosphorothiate chimeras: thermal stability and antisense inhibition of gene expression" Nucleic Acids Research, 25( 16), pp. 3310-3317. Borecky, L.,et a!. (1981-1982) "Therapeutic use of double-stranded RNAs in man" Tex. Rep. Bioi. Med., 41, pp. 575-581. Cameron, F. H., eta!. (1994) "Multiple domains in a ribozyme con­struct confer increased suppresive activity in monkey cells" Antisense Research and Development, 4(2), pp. 87-94. Cameron, F. H., Jennings, P. A., (1991) "Inhibition of gene expres­sion by a short sense fragment" Nucleic Acids Research, 19(3), pp. 469-475. Cohli, H., eta!. ( 1994) "Inhibition ofHIV-1 multiplication in a human CD4+ lymphocytic cell line expressing antisense and sense RNA molecules containing HIV-1 packaging signal and Rev response ele­ment(s)" Antisense Research and Development, 4(1), pp. 19-26. Coleman, J., et a!. (1984) "The use of RNAs complementary to specific mRNAs to regulate the expression of individual bacterial genes" Cell, 37(2), pp. 429-436. Dolnick, B. J. (1997) "Naturally occuring antisense RNA" Pharm. Ther. 75(3), pp. 179-184.

Page 6: 112   andrew fire - 7622633 - genetic inhibition by double-stranded rna

US 7,622,633 B2 Page 6

Fire, A., eta!. (1991) "Production of antisense RNA leads to effective and specific inhibition of gene expression in C. elegans muscle" Development, 113(2), pp. 503-514. Flavell, R. B., (1994) "Inactivation of gene expression in plants as a consequence of specific sequence duplication" Proc. Nat!. Acad. Sci. US A, 91(9), pp. 3490-3496. Good, P. D., eta!. (1997) "Expression of small, therapeutic RNAs in human cell nuclei" Gene Ther. 4(1), pp. 45-54. Graham, G. J. et a!. (1990) "RNA transcripts of the human immunodeficiency virus transactivation response element can inhibit action of the viral transactivator" Proc. Nat!. A cad. Sci. US A, 87( 15), pp. 5817-5821. Ha, I., Wightman, B., et al. (1996) "A bulged lin-4/lin-14 RNA duplex is sufficient for Caenorhabditis elegans lin-14 temporal gra­dient formation" Genes Dev., 10(23), pp. 3041-3050. James, W. (1991) "Towards gene-inhibition therapy: a review of progress and prospects in the field of antiviral antisense nucleic acids and ribozymes" Antiviral Chern & Chemother. 2(4), pp. 191-214. Kibler, K. V., eta!, (1997) "Double-stranded RNA is a trigger for apoptosis in vaccinia virus-infected cells" Journal ofVirology, 71(3), pp. 1992-2003. Klaff, P., eta!. (1996) "RNA structure and the regulation of gene expression" Plant Mol. Bioi. 32( 1-2), pp. 89-106. Liebhaber, S. A., eta!. (1992) "Translation inhibition by an mRNA coding region secondary structure is determined by its proximity to the AUG initiation codon" J. Mol. Bioi. 226(3), pp. 609-621. Loomis, W. F., et al. ( 1991) "Antisense RNA inhibition of expression of a pair of tandemly repeated genes results in a delay in cell-cell adhesion in Dictyostelium" Antisense Res. Dev. 1(3), pp. 255-260. Manche, L., et a!. (1992) "Interactions between double-stranded RNA regulators and the protein kinase DAl" Molecular and Cellular Biology, 12(11), pp. 5238-5248. Matze, M. A., et a!. (1995) "How and why do plants inactivate homologous (trans) genes?" Plant Physiol. 107(3), pp. 679-685. Mercola, D., et al. (1995) "Antisense approaches to cancer gene therapy" Cancer Gene Ther. 2(1), pp. 47-59. Mikoshiba, K., eta!. (1991) "Molecular biology of myelin basic protein: gene rearrangement and expression of anti-sense RNA in myelin-deficient mutants" Comp. Biochem. Physiol. C., 98(1), pp. 51-61. Okano, H., eta!. (1991) "Myelin basic protein gene and the function of antisense RNA in its repression in myelin-deficient mutant mouse" J. Neurochem., 56(2), pp. 560-567. Podivinsky, E., Walton, E. F., et al. (1994) "Extraction of RNA from kiwi fruit tissues" Biotechniques, 16(3), pp. 396-398. Sarver, N., et al. (1990) "Ribozymes as potential anti-HIV-1 thera­peutics agents" Science, 247(4947), pp. 1222-1225. Sullenger, B. A., eta!. (1990) "Expression of chimeric tRNA-driven antisense transcripts renders NIH 3T3 cells highly resistant to moloney murine leukemia virus replication" Mol. Cell. Bioi. 10(12), pp. 6512-6523. Sullenger, B. A., eta!. (1993) "Tethering ribozymes to a retroviral packaging signal for destruction of viral RNA" Science, 262(5139), pp. 1566-1569. Uhlmann, E., eta!. (1990) "Antisense oligonucleotides: a new thera­peutic principle" Chemical Reviews, 9(4), pp. 544-584. Yamamoto, R., et al. (1997) "Inhibition of transcription by the TAR RNA of HIV-1 in a nuclear extract of HeLa cells" Nucleic Acids Research, 25(17), pp. 3445-3450. Parrish et a!. "Functional anatomy of a dsRNA trigger: Differential requirements for the the trigger strands in RNA interference" Mol. Cell (2000) 6: 1077-1087. Elbashir et al. "RNA interference is mediated by 21- and 22-nucleotide RNA" Genes Dev. (2001) 15: 188-200. Elbashir eta!. "Duplexes of21-nucleotide RNAs mediate RNA inter­ference in cultured manunalian cells" Nature (200 1) 411: 494-498. Bass "The short answer" Nature (2001). Davenport "A faster way to shut down genes" Science (200 1) 292:1469-1471. Hutvagner et a!. "A cellular function for the RNA-interference enzyme Dicer in the maturation of the let-7 small temporal RNA" Science (2001) 294: 834-838.

Grishok et a!. "Genes and mechanisms related to RNA interference regulate expression of the small temporal RNAs that control C. elegans developmental timing" Cell (2001) 105: 23-44. Caplen et al. "Specific inhibition of gene expression by small double­stranded RNAs in invertebrate and vertebrate systemss" Proc Nat! A cad Sci USA (200 1) early edition pnas 171251798 pp. 1-6. Baker eta!., "RNAi of the receptor tyrosine phosphatase HmLAR2 in a single cell of an intact leech embryo leads to growth-cone collapse," Current Biology (2000) vol. 10, pp. 1071-1074. Bass, "Double-stranded RNA as a template for gene silencing," Cell (2000) vol. 101, pp. 235-238. Bastin et a!, "Flagellum ontogeny in trypanosomes studied via an inherited and regulated RNA interference sytem," J Cell Science (2000) vol. 113, pp. 3321-3328. Baulcombe, "Unwinding RNA silencing," Science (2000) vol. 290, pp. 1108-1109. Bhat eta!., "Discs Lost, a novel multi-PDZ domain protein, estab­lishes and maintains epithelial polarity," Cell (1999), vol. 96, pp. 893-845. Bosher eta!., "RNA interference: genetic wand and genetic watch­dog," Nature Cell Biology (2000) vol. 2, pp. E31-E36. Caplen eta!, "dsRNA-mediated gen silencing in cultured Drosophila cells: a tissue culture model for the analysis of RNA interference," Gene (2000), vol. 252, pp. 95-105. Chuang et at. "Specific and heritable genetic interference by double­stranded RNA in Arabidopsis thaliana," PNAS (2000) vol. 97, pp. 4985-4990. Colussi eta!., "Debcl, a proapoptotic Bcl-2 homologue, is a compo­nent of the Drosophila melanogaster cell death machinery," J Cell Biology (2000) vol. 148, pp. 703-714. Denef et a!., "Hedgehog induces opposite changes in turnover and subcellular localization of Patched and Smoothened," Cell (2000) vol. 102, pp. 521-531. Domeier et al., "A link between RNA interference and nonsense­mediated decay in Caenorhabditis elegans ," Science (2000) vol. 289, pp. 1928-1930. Fagard et a!., "AGO!, ODE-2, and RDE-1 are related proteins requried for post-transcriptional gene silencing in plants, quelling in fungi, and RNA interference in animals," PNAS (2000) vol. 97, pp. 11650-11654. Fortier et a!., "Temperature-dependent gene silencing by an expressed inverted repeat in Drosphila," Genesis (2000) vol. 26, pp. 240-244. Fraser et a!., "Functional genomic analysis of C. elegans chromo­some I by systematic RNA interference," Nature (2000) vol. 408, pp. 325-330. Hill et al., "dpy-18 Encodes an a-subunit of prolyl-4-hydrolase in Caenorhabditis elegans," Genetics (2000) vol. 155, pp. 1139-1148. Hsieh et a!., "The RING finger/B-Box factor TAM-1 and a retinoblastoma-like protein LIN-35 modulate context-dependent gene silencing in Caenorhabditis elegans," Genes & Development (1999) vol. 13, pp. 2958-2970. Huang eta!., "The proneural gene amos promotes multiple dendritic neuron formation in the Drosophila peripheral nervous system," Neuron (2000) vol. 25, pp. 57-67. Hughes eta!., "RNAi analysis of Deformed, proboscipedia and Sex combs reduced in the milkweed bug Oncopeitus fasciatus: novel roles for Hox genes in the Hemipteran head," Development (2000) vol. 127, pp. 3683-3694. Hunter, "Gene silencing: Shrinking the black box ofRNAi," Current Biology (2000) vol. 10, pp. Rl37-Rl40. Kennerdell et a!., "Heritable gene silencing in Drosphila using double-stranded RNA," Nature Biotechnology (2000) vol. 17, pp. 896-898. Kim et al., "Positioning of longitudinal nerves in C. elegans by nidogen," Science (2000) vol. 288, pp. 150-154. Kostich eta!., "Identification and molecular-genetic characterization of a LAMP/CD68-like protein from Caenorhabditis elegans," J Cell Science (2000) vol. 113, pp. 2595-2606. Lam et al., "Inducible expression of double-stranded RNA directs specific genetic interference in Drosphila," Current Biology (2000) vol. 10, pp. 957-963.

Page 7: 112   andrew fire - 7622633 - genetic inhibition by double-stranded rna

US 7,622,633 B2 Page 7

Lewis eta!., "Distinct roles of the homeotic genes Ubx and abd-A in beetle embryonic abdonimal appendage development," PNAS (2000) vol. 97, pp. 4504-4509. Liu eta!, "Overlapping roles of two Hox genes and the exd ortholog cah-20 in diversification of the C. elegans postembryonic meso­derm," Development (2000) vol. 127, pp. 5179-5190. Liu eta!., "Essential roles for Caenorhabditis elegans lamin gene in nuclear organization, cell cycle progression, and spatial organization of nuclear pore complexes," Molecular Biology Cell (2000) vol. 11, pp. 3937-3947. Lohmann eta!., "Silencing of developmental genes in Hydra," Devel­opmental Biology (1999) vol. 214, pp. 211-214. Maine, "A conserved mechanism for post-transcriptional gene silencing?" Genome Biology (2000) vol. I, pp. 1018.1-1018.4. Mette et a!., "Transcriptonal silencing and promoter methylation triggered by double-stranded RNA," The EMBO Journal (2000) vol. 19, pp. 5194-6201. Marx, "Interfering with gene expression," Science (2000) vol. 288, pp. 1370-1372. Melendez et al., "Caenorhabditis elegans lin-13, a member of the LIN -35 Rb class of genes involved in vulvar development, encodes a protein with zinc fingers and an LXCXE motif," Genetics (2000) vol. 155, pp. 1127-1137. Nakano et al., "RNA interference for the organizer-specific gene Xlim-1 in Xenopus embryos," Biochemical Biophysical Research Communications (2000) vol. 274, pp. 434-439. Oates et aL. "Too much interference: Injection of double-stranded RNA has nonspecific effects in the zebrafish embryo," Developmen­tal Biology (2000) vol. 224, pp. 20-28. Oelgeschliiger et a!., "The evolutionarily conserved BMP-binding protein Twisted gastrulation promotes BMP signalling," Nature (2000) vol. 405, pp. 757-763. Parrish eta!., "Functional anatomy of a dsRNA trigger: Differential requirement for the two trigger strands in RNA interference," Molecular Cell (2000) vol. 6, pp. 1077-1087. Pichler et al., "OOC-3, a novel putative transmembrane protein required for establishment of cortical domains and spindle orienta­tion in the Ps blastomere of C. elegans embryos," Development (2000) vol. 127, pp. 2083-2073. Pineda eta!., "Searching for the prototypic eye genetic network: Sine oculis is essential for eye regeneration in planarians," PNAS (2000) vol. 97, pp. 4525-4529. Sawa at a!., "Components of the SWI/SNF complex are required for asymmetric cell division in C. elegans," Molecular Cell (2000) vol. 6, pp. 617-624. Shi eta!., "Genetic interference in Trypanosoma brucei by heritable and inducible double-stranded RNA," RNA (2000) vol. 6, pp. 1069-1078. Shippy et al., "Analysis of maxillopedia expression pattern and larval cuticular phenotype in wild-type and mutant Tribolium," Genetics (2000) vol. 155, pp. 721-731. Stauber et al., "Function of bicoid and hunchback homologs in the basel cyclorhaphan fly Megeselia (Phoridae)," PNAS (2000) vol. 97, pp. 10844-10849. Svoboda eta!., "Selective-reduction of dormant maternal mRNAs in mouse oocytes by RNA interference," Development (2000) vol. 127, pp. 4147-4156. Tavernarakis eta!., "Heritable and inducible genetic interference by double-stranded RNA encoded by transgenes," Nature Genetics (2000) vol. 24, pp. 180-183. Ui-Tei eta!., "Sensitive assay of RNA interference in Drosphilia and Chinese hamster cultured cells using firefly luciferase gene as target," FEBS Letters (2000) vol. 479, pp. 79-82. Wang eta!., "Inhibition of Trypanosoma brucei gene expression by RNA interference using an integratable vector with opposing T7 promoters," J Biological Chemistry (2000) electronically published as Manuscript M008405200, pp. 1-30 and Figures 1-6. Willert et al., "A Drosphila Axin homolog. Daxin, inhibits Wnt sig­nalling," Development (1999) vol. 126, pp. 4165-4173. Wu-Scharf et a!., "Transgene and transposon silencing in Chlamydomonas reinhardtii by a DEAH-box RNA helicase," Sci­ence (2000) vol. 290, pp. 1159-1162.

Yang et al., "Evidence that processed small dsRNAs may mediate sequence-specific mRNA degradation during RNAi in Drosphila embryos," Current Biology (2000) vol. 10, pp. 1191-1200. Zamore et al, "RNAi: Double-stranded RNA directs the ATP-depen­dent cleavage of mRNA at 21 to 23 nucleotide intervals," Cell (2000) vol. 101, pp. 25-33. Grishok et al., "Genetic Requirements for Inheritance ofRNAi in C. elegans", Science, vol. 287, Mar. 31, 2000, pp. 2494-2497. Sharp et al., "RNA Interference", Science, vol. 287. Mar. 31, 2000, pp. 2431 and 2433. Samuel E. Driver, Gregory S. Robinson, Jean Flanagan, Wei Shen, Lois E.H. Smith, David W. Thomas, and Peter C. Roberts, "Oligonucleotide-based inhibition of embryonic gene expression," Nature Biotechnology, vol. 17, Dec. 1999, pp. 1184-1187. Alejandro Sanchez Alvarado and Phillip A. Newmark, "Double­stranded RNA specifically disrupts gene expression during planarian regeneration," Proc. Nat!. Acad. Sci. USA, vol. 96, Apr. 1999, Devel­opmental Biology, pp. 5049-5054. James D. Thompson, "Shortcuts from gene sequence to function," Nature Biotechnology, vol. 17, Dec. 1999, pp. 1158-1159. RatcliffF eta!: "A similarity between viral defense and gne silencing in plants", Science, vol. 276, No. 93, Jun. 6, 1997, pp. 1558-1560, XP002095874, see the whole document. Fire, A. et a!., "Production of antisense RNA leads to effective and specific inhibition of gene expression in C. elegans muscle", Devel­opment (Cambridge, UK) (1991), 113(2), 503-14 XP002103600 cited in the application, seep. 508, right-hand colunm, paragraph 2, seep. 509, right-hand column- p. 511, right-handcolumn, seep. 512, 'Discussion' and figure 7. Matzke M A et a!: "How and Why Do Plants Inactive Homologous (Trans)Genes?",PlantPhysiology,vol.107,No.3,Mar.1, 1995,pp. 679-685, XP002021174, seep. 608, left-hand colunm, paragraph 3 -right-hand column, paragraph 1, seep. 682. Fire A et a!: "Potent and specific genetic interference in double -stranded RNA in caenorhabditis elegans", Nature, (Feb. 19, 1998) 391 (6669) 806-11., XP002095876, cited in the application, see the whole document. Montgomery M K et a!: "Double stranded RNA as a mediator in sequence-specific genetic silencing and co-specific genetic silencing and co- suppression", Trends in Genetics, (Jul. 1998) 14 (7) 255-8., XP004124680, cited in the application, see the whole document. Timmons Let a!: "Specific interference by ingested dsRNA", Nature, (Oct. 29, 1998) 395 (6705) 854., XP002103601, cited in the appli­cation, see the whole document. Baum eta!., "Inhibition of protein synehtsis in reticulocyte lysates by a double-stranded RNA component in HeLa mRNA" Biochem Biophys Res Comm, Jul. 18, 1983, 114:41-49. Pratt eta!., "Regulatin of in vitro translation by double-stranded RNA in mammalian cell mRNA preparations" Nucl Acids Res, Feb. 25, 1988, 16:3497-3510. Klaff eta!., "RNA structure and the regulation fo gene expression" Plant Mol Bioi. 1996, 32:89-106. Doinick, "Natural! occurring antisense RNA" Pharmacol Ther. 1997, 75:179-184. Wagner eta!., "Double-stranded RNA poses puzzle" Nature, Feb. 19, 1998, 391:744-745. Harbinder et a!., "Genetically targeted cell disruption in Caenorhabditis elegans" Proc Nat! Acad Sci USA, Nov. 1997, 94:13128-131133. Harcourt et al., "Ebola virus inhibits induction of genes by double­stranded RNA in endothelial cells" Virology, 1998, 252:179-188. Kumar and Carmichael, "Antisense RNA: Function and fate of duplex RNA in cells of higher eukaryotes" Micro bioi Mol Bioi Rev, Dec. 1988,62:1415-1434. Suzuki eta!., "Activation of target-tissue immune-recognition mol­ecules by double-stranded polynucleotides" Proc Nat! Acad Sci USA, Mar. 1999, 96:2285-2290. Hunter, "A touch of elegance with RNAi" Curr Bioi, Jun. 17, 1999, 9:R440-R442. Tuschi et al., "Targeted mRNA degradation by double-stranded RNA in vitro" Genes Dev. Dec. 15, 1999, 13:3191-3197. Fire, "RNA-triggered gene silencing" Trends Genet, Sep. 1999, 15:358-363.

Page 8: 112   andrew fire - 7622633 - genetic inhibition by double-stranded rna

US 7,622,633 B2 Page 8

Tabara et al., "The rde-1 Gene, RNA Interference, and transposon silencing in C. elegans" Cell, Oct. 15, 1999,99:123-132. Ketting eta!., "mut-7 of C. elegans, required fortransposon silencing and RNA interference, is a homolog of Werner syndrome helicase and RNaseD" Cell, Oct. 15, 1999, 99:133-141. Bosher et a!., "RNA interference can target pre-mRNA: Conse­quences for gene expression in a Caenorhabditis elegans operon" Genetics, Nov. 1999, 153:1245-1256. Thompson, "Shortcuts from gene Sequence to function" Nature Biotechnology, Dec. 199, 17:1158-1159. Driver et al., "Oligonucleotide-based inhibition of embryonic gene expression" Nature Biotechnology, Dec. 1999, 17:1184-1187. Wargelius et a!., "Double-stranded RNA induces specific develop­mental defects in zebrafish embryos" Biochem Biophys Res Comm, 1999,263:156-161. Li at a!., "Double-stranded RNA infection produces null phenotypes in zebrafish" Developmental Biology, Jan. 15, 2000, 217:394-405. Wlanny et a!., "Specific interference with gene function by double­stranded RNA in early mouse development" Nature Cell Biology, Feb. 2000, 2:70-75. Catalanotto et al., "Gene silencing in worms and fungi" Nature, Mar. 16, 2000,404:245. Hammond et al., "An RNA-directed nuclease mediates post-tran­scriptional gene silencing in Drosophila cells" Nature, Mar. 16, 2000, 404:293-296. Ketting and Plasterk, "A genetic link between co-suppression and RNA interference in C. elegans" Nature, Mar. 16, 2000, 404:296-298. Fire, A., eta!., (1998) Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans. Nature 391: 806-811. Fire, A., (1998) "RNAi info from the Fire Lab" (Web Document with protocols to accompany the Fire eta!., Nature paper). Posted on our public-access web site from Feb. 1, 1998. Fire, A., and Fleenor, J. (1998) On the generality of RNA-mediated interference. Worm Breeder's Gazette 15(3): 8 (Jun. 1, 1998). Montgomery, M., and Fire, A. (1998) Double-stranded RNA as a mediator in sequence-specific genetic silencing and co-suppression. Trends in Genetics 14: 255-258. Harfa, B., eta!. (1998) Analysis of a Caenorhabditis elegarts twist homolog identifies conserved and divergent aspects of mesodermal patterning. Genes and Development 12: 2623.2636. Tabara, H., eta!. (1998) RNAi in C. elegans: Soaking in the genome sequence. Science 282: 430-431. Timmons, L., and Fire, A. ( 1998) Specific interference by ingested dsRNA. Nature, 395: 854. Montgomery, M.K., eta!., ( 1998) RNA as a target of double-stranded RNA-mediated genetic interference in Caenorhabditis elegan. Prod. Nat!. Acad. Sci. 95:15502-15507.

Fire, A., eta!., (1991) Production of antisense RNA leads to effective and specific inhibition of gene expression in C. elegans muscle. Development 113: 503-514. Grieson, D. et a!., (1991) Does co-suppression of sense genes in transgenic plants involve antisense RNA? Trends in Biotechnology 9:122-123. Nellen, W., and Lichtenstein, C. (1993) What makes an mRNA anti­sensitive? Trends in Biochemical Sciences 18: 419-423. Guo, S. and Kemphues, K (1995) par-1, a gene required for estab­lishing polarity in. C. elegans embryos, encodes a putative Ser/Thr kinase that is asymmetrically distributed. Cell81: 611-620. Matzke, M.A., and Matzke, A.J.M. (1995) How and why do plants inactivate homologous (trans )genes? Plant Physiology 107: 679-685. Metziaff, M., et a!., (1997) RNA-mediated RNA degradation and chalcone synthetase A silencing in petunia. Cell 88: 845-854. Ratfliff, F., et al., (1997) A similarity between viral defense and gene silencing in plants. Science 276: 1588-1560. Jorgensen, R.A., et a!., (1998) An RNA-based information super­highway in plants. Science 279: 1486-1487. Waterhouse, P.M., et al., ( 1998) Virus resistance and gene silencing in plants can be induced by simultaneous expression of sense and antisense RNA. Proc. Nat!. Acad. Sci. 95: 13959-13964. Ngo, H., eta!., (1998) Double-stranded RNA induces mRNA degra­dation in Trypanosoma bruce!. Proc. Nat!. Acad. Sci. 95: 14687-14692. Kennerdell, J., and Carthew, R. (1998) Drosophila frizzled and frizzled2 act in the wingless pathway as determined by dsRNA­medication genetic interference. Cell95: 1017-1026. Jorgensen, R.A., eta!., (1999) Do unintended artisense transcripts contribute to sense-co-suppression in plants? Trends in Genetics 15: 11-12. Misquitta, L, and Paterson, B.M. (1999) Targeted disruption of gene function in Drosophila by RNa interference.(RNA-I): A role for nautilus in embryonic somatic muscle formation. Proc. Nat!. Acad. Sci. 96: 1451-1456. Sharp, P.A. (1999) RNAi and double-stranded RNA. Gene and Development 13: 139-141. Seydoux, G., et a!. (1996) Repression of gene expression in the embryonic germ lineage of C. elegans, Nature 382: 713-716. Starn, M., eta!. (1997) The silence of genes in transgenic plants. annals of Botany 79: 3-12. Nellen, W., and Lichtenstein, C. (1993) What makes an mRNA anti­sense-itive? Trends in Biochemical Sciences 18: 419-423. Proud, C.G. (1995) PKR: A new name and new roles. Trends in Biochemical Sciences 20: 241-246. Jacobs, B.L., and Langland, J.O. (1996) When two strands are better than one: The mediators and modulators of the cellular response to double-stranded RNA Virology 219: 339-349.

* cited by examiner

Page 9: 112   andrew fire - 7622633 - genetic inhibition by double-stranded rna

c 8 5.0kb

unc-22 I II m ml : II Ill II I i: om = I II[Zi 00

A B A 1.0kb =

Unc-54 . rp:;;J fjill:l c 13 00

FIG. 1 gfp fusions 1.0kb

:::==

NUCLEAR

MITOCHONDRIAL

~ rJ1 • ""0 ~

""""' ~ = """"'

~ 0 ~ t-.J ~~ t-.J 0 0 \0

rF1 ::r ('D ('D ...... ..... 0 ..... (.II

Lj rJl ......:J ~ N N ~ w w o= N

Page 10: 112   andrew fire - 7622633 - genetic inhibition by double-stranded rna

~ :::::>

~ H

!::J :;:)

~

CONTROL RNA (ds-unc22a) ---CONTROL RNA(ds-unc22a)

,.

.... ,. .. ,~··;:_~·~!~•A.

FIG. 28 CONTROL RNA ( ds-unc22a)

FIG. 2C

....-

..J

~ ::::> 0 <{

~ ::::> Cl <(

H

ds-gfpG RNA

FIG. 20 ds-gfpG RNA

)~

FIG. 2E ds-gfpG RNA

FIG. 2F

..... _.

!::; ::J 0 <(

!:J ::J Cl <(

ds-lacZL RNA

·---;-.... .-.• -...... !'-' .. -~·--- -.. :·:~~-:-::~;;;::·;· ;:··~:tfii~Mt:

~ l

FIG. 2G ds-/acZL RNA ·.·:),;~·;.:.·

· .... _: ·-:;-:~:~

H ' ,: j<

FIG. 2H ds-lacZL RNA

FIG. 21

~ 00 • ~ ~ ~ ~ = ~

z 0 ~ N ~ ... N 0 0 \0

rFJ

=­('D ('D ..... N 0 ..... Ul

d rJl -....l 0.., N N 0.., w w

= N

Page 11: 112   andrew fire - 7622633 - genetic inhibition by double-stranded rna

U.S. Patent Nov. 24, 2009 Sheet 3 of 5

. . ,, .

! • ••• ' •• •' •.

.. . . . .. ' .. -. . . . .~ · ... • '•' . : '· · ... '•' . . .

FIG. 3A FIG. 38

FIG. 3C FIG. 30

US 7,622,633 B2

Page 12: 112   andrew fire - 7622633 - genetic inhibition by double-stranded rna

--------~---ds~A------------30,000

(.1)

~ ~

1.00 1.800,000

0.75-1"1['

u= 0.50-g, z: 0 .-= ~ u.. 0.25 .J! ...

600,000 m~ 70,000

'''ill•

~. ·~ ·n:n: ... ,,.

•t•,······

····•r•lr.""'' Ill ••·••·

-'ANTISENSE'- 'SENSE' CONTROL ....._RNA 3,600,000 1,200,000 3,600,000 3,600,000 ..._MOLECULES I II II II II I I II II IIIII ill II II ill 1111111111 ---, INJECTED

PER GONAD (APPRO X)

FIG.4A

0 00 .t!UUifiltLJJUkllilhl Hl1tii:JD liJIJLRIN W"IHIIII llklllllll Nllllllll fiJI II II II I I II II !LILL.-J

. 12345 12345 12345 12345 12345 12345 12345 12345 12345

WILD TYPE 100% 75% 66% 50~ 33% 25%

SEVERLY AFFECTED O%-

FIG. 48

~ 7J). • ~ ~ ~ ~ = ~

z 0 ~ N ~ ... N 0 0 \0

rFJ

=­('D ('D ...... ... 0 ...... Ul

d rJl -....l 0... N N 0... w w

= N

Page 13: 112   andrew fire - 7622633 - genetic inhibition by double-stranded rna

BACTERIAL CELL WALL

FIG. 5A

PD4251 WORMS

FIG. 58

PD4251 WORMS FED BACTERIA EXPRESSING gfp dsRNA

FIG. 5C

~ 00 • ""d ~ ~ ~ = ~

z 0 ~ N ~ ... N 0 0 \0

rFJ

=­('D ('D ..... Ul 0 ..... Ul

d rJl

"'--...1 0'1 N

"'N 0'1 w w

= N

Page 14: 112   andrew fire - 7622633 - genetic inhibition by double-stranded rna

US 7,622,633 B2 1

GENETIC INHIBITION BY DOUBLE-STRANDED RNA

RELATED APPLICATION

This application is a divisional application of application Ser. No. 09/215,257, filed Dec. 18, 1998, now U.S. Pat. No. 6,506,559 which claims the benefit ofU.S. Provisional Appli­cation Ser. No. 60/068,562, filed Dec, 23, 1997.

GOVERNMENT RIGHTS

This invention was made with U.S. government support under grant numbers GM-37706, GM-17164, HD-33769 and GM-07231 awarded by the National Institutes of Health. The U.S. government has certain rights in the invention.

BACKGROUND OF THE INVENTION

1. Field of the Invention

The present invention relates to gene-specific inhibition of gene expression by double-stranded ribonucleic acid (dsRNA).

2. Description of the Related Art

Targeted inhibition of gene expression has been a long-felt need in biotechnology and genetic engineering. Although a major investment of effort has been made to achieve this goal, a more comprehensive solution to this problem was still needed.

Classical genetic techniques have been used to isolate mutant organisms with reduced expression of selected genes. Although valuable, such techniques require laborious mutagenesis and screening programs, are limited to organ­isms in which genetic manipulation is well established (e.g., the existence of selectable markers, the ability to control genetic segregation and sexual reproduction), and are limited

2 Use of Antisense Nucleic Acids to Engineer Interference

Antisense technology has been the most commonly described approach in protocols to achieve gene-specific interference. For antisense strategies, stochiometric amounts of single-stranded nucleic acid complementary to the mes­senger RNA for the gene of interest are introduced into the cell. Some difficulties with antisense-based approaches relate to delivery, stability, and dose requirements. In general, cells do not have an uptake mechanism for single-stranded nucleic

10 acids, hence uptake of unmodified single-stranded material is extremely inefficient. While waiting for uptake into cells, the single-stranded material is subject to degradation. Because antisense interference requires that the interfering material accumulate at a relatively high concentration (at or above the

15 concentration of endogenous mRNA), the amount required to be delivered is a major constraint on efficacy. As a conse­quence, much of the effort in developing antisense technol­ogy has been focused on the production of modified nucleic acids that are both stable to nuclease digestion and able to

20 diffuse readily into cells. The use of antisense interference for gene therapy or other whole-organism applications has been limited by the large amounts of oligonucleotide that need to be synthesized from non-natural analogs, the cost of such synthesis, and the difficulty even with high doses of main-

25 taining a sufficiently concentrated and uniform pool of inter­fering material in each cell.

Triple-Helix Approaches to Engineer Interference A second, proposed method for engineered interference is

based on a triple helical nucleic acid structure. This approach 30 relies on the rare ability of certain nucleic acid populations to

adopt a triple-stranded structure. Under physiological condi­tions, nucleic acids are virtually all single- or double­stranded, and rarely if ever form triple-stranded structures. It has been known for some time, however, that certain simple

35 purine- or pyrimidine-rich sequences could form a triple­stranded molecule in vitro under extreme conditions of pH (i.e., in a test tube). Such structures are generally very tran­sient under physiological conditions, so that simple delivery of unmodified nucleic acids designed to produce triple-strand

40 structures does not yield interference. As with antisense, development of triple-strand technology for use in vivo has focused on the development of modified nucleic acids that would be more stable and more readily absorbed by cells in vivo. An additional goal in developing this technology has

to applications in which a large number of cells or organisms can be sacrificed to isolate the desired mutation. Even under these circumstances, classical genetic techniques can fail to produce mutations in specific target genes of interest, particu­larly when complex genetic pathways are involved. Many applications of molecular genetics require the ability to go beyond classical genetic screening techniques and efficiently produce a directed change in gene expression in a specified group of cells or organisms. Some such applications are knowledge-based projects in which it is of importance to understand what effects the loss of a specific gene product (or 50 products) will have on the behavior of the cell or organism. Other applications are engineering based, for example: cases

45 been to produce modified nucleic acids for which the forma­tion of triple-stranded material proceeds effectively at physi­ological pH.

Co-Suppression Phenomena and Their Use in Genetic Engi­neering

A third approach to gene-specific interference is a set of operational procedures grouped under the name "co-suppres­sion". This approach was first described in plants and refers to the ability of transgenes to cause silencing of an unlinked but homologous gene. More recently, phenomena similar to co­suppression have been reported in two animals: C. elegans and Drosophila. Co-suppression was first observed by acci­dent, with reports coming from groups using transgenes in attempts to achieve over-expression of a potentially useful locus. In some cases the over-expression was successful while, in many others, the result was opposite from that expected. In those cases, the transgenic plants actually showed less expression of the endogenous gene. Several mechanisms have so far been proposed for transgene-medi­ated co-suppression in plants; all of these mechanistic pro­posals remain hypothetical, and no definitive mechanistic description of the process has been presented. The models that have been proposed to explain co-suppression can be

in which is important to produce a population of cells or organisms in which a specific gene product (or products) has been reduced or removed. A further class of applications is 55 therapeutically based in which it would be valuable for a functioning organism (e. g., a human) to reduce or remove the amount of a specified gene product (or products). Another class of applications provides a disease model in which a physiological function in a living organism is genetically 60 manipulated to reduce or remove a specific gene product (or products) without making a permanent change in the organ­ism's genome.

In the last few years, advances in nucleic acid chemistry and gene transfer have inspired new approaches to engineer 65

specific interference with gene expression. These approaches are described below.

Page 15: 112   andrew fire - 7622633 - genetic inhibition by double-stranded rna

US 7,622,633 B2 3

placed in two different categories. In one set of proposals, a direct physical interaction at the DNA- or chromatin-level between two different chromosomal sites has been hypoth­esized to occur; an as-yet-unidentified mechanism would then lead to de novo methylation and subsequent suppression

4

of gene expression. Alternatively, some have postulated an RNA intermediate, synthesized at the trans gene locus, which might then act to produce interference with the endogenous gene. The characteristics of the interfering RNA, as well as the nature of the interference process, have not been deter­mined. Recently, a set of experiments with RNA viruses have provided some support for the possibility of RNA intermedi­ates in the interference process. In these experiments, a rep­licating RNA virus is modified to include a segment from a gene of interest. This modified virus is then tested for its ability to interfere with expression of the endogenous gene. Initial results with this technique have been encouraging, however, the properties of the viral RNA that are responsible for interference effects have not been determined and, in any case, would be limited to plants which are hosts of the plant 20

certain trans genes can cause interference (i.e., a quantitative decrease in the activity of the corresponding endogenous locus) but that most trans genes do not produce such an effect. The lack of a predictable effect in plants, nematodes, and insects greatly limits the usefulness of simply adding trans­genes to the genome to interfere with gene expression. Viral­mediated co-suppression in plants appears to be quite effec­tive, but has a number of drawbacks. First, it is not clear what aspects of the viral structure are critical for the observed

10 interference. Extension to another system would require dis­covery of a virus in that system which would have these properties, and such a library of useful viral agents are not available for many organisms. Second, the use of a replicating

15 virus within an organism to effect genetic changes (e.g., long­or short-term gene therapy) requires considerably more monitoring and oversight for deleterious effects than the use of a defined nucleic acid as in the present invention.

VIruS.

Distinction between the Present Invention and Antisense Approaches

The present invention differs from antisense-mediated 25

interference in both approach and effectiveness. Antisense­mediated genetic interference methods have a major chal­lenge: delivery to the cell interior of specific single-stranded nucleic acid molecules at a concentration that is equal to or greater than the concentration of endogenous mRNA.

30 Double-stranded RNA-mediated inhibition has advantages both in the stability of the material to be delivered and the concentration required for effective inhibition. Below, we disclose that in the model organism C. elegans, the present invention is at least 100-fold more effective than an equiva-

35 lent antisense approach (i.e., dsRNA is at least 100-foldmore effective than the injection of purified antisense RNA in reducing gene expression). These comparisons also demon­strate that inhibition by double-stranded RNA must occur by

The present invention avoids the disadvantages of the pre­viously-described methods for genetic interference. Several advantages of the present invention are discussed below, but nnmerous others will be apparent to one of ordinary skill in the biotechnology and genetic engineering arts.

SUMMARY OF THE INVENTION

A process is provided for inhibiting expression of a target gene in a cell. The process comprises introduction of RNA with partial or fully double-stranded character into the cell or into the extracellular environment. Inhibition is specific in that a nucleotide sequence from a portion of the target gene is chosen to produce inhibitory RNA. We disclose that this process is (1) effective in producing inhibition of gene expres­sion, (2) specific to the targeted gene, and (3) general in allowing inhibition of many different types of target gene.

The target gene may be a gene derived from the cell, an endogenous gene, a trans gene, or a gene of a pathogen which is present in the cell after infection thereof. Depending on the

a mechanism distinct from antisense interference.

Distinction between the Present Invention and Triple-Helix Approaches

40 particular target gene and the dose of double stranded RNA material delivered, the procedure may provide partial or com­plete loss of function for the target gene. A reduction or loss of gene expression in at least 99% of targeted cells has been shown. Lower doses of injected material and longer times

The limited data on triple strand formation argues against the involvement of a stable triple-strand intermediate in the present invention. Triple-strand structures occur rarely, if at all, under physiological conditions and are limited to very unusual base sequence with long runs of purines and pyrim­idines. By contrast, dsRNA-mediated inhibition occurs effi­ciently under physiological conditions, and occurs with a wide variety of inhibitory and target nucleotide sequences. 50

The present invention has been used to inhibit expression of 18 different genes, providing phenocopies of null mutations

45 after administration of dsRNA may result in inhibition in a smaller fraction of cells. Quantitation of gene expression in a cell may show similar amounts of inhibition at the level of accnmulation of target mRNA or translation of target protein.

in these genes of known function. The extreme environmental and sequence constraints on triple-helix formation make it unlikely that dsRNA-mediated inhibition in C. elegans is 55

mediated by a triple-strand structure.

The RNA may comprise one or more strands of polymer­ized ribonucleotide; it may include modifications to either the phosphate-sugar backbone or the nucleoside. The double­stranded structure may be formed by a single self-comple­mentary RNA strand or two complementary RNA strands. RNA duplex formation may be initiated either inside or out­side the cell. The RNA may be introduced in an amount which allows delivery of at least one copy per cell. Higher doses of double-stranded material may yield more effective inhibition. Inhibition is sequence-specific in that nucleotide sequences corresponding to the duplex region of the RNA are targeted

Distinction between Present Invention and Co-Suppression Approaches

The trans gene-mediated genetic interference phenomenon called co-suppression may include a wide variety of different processes. From the viewpoint of application to other types of organisms, the co-suppression phenomenon in plants is dif­ficult to extend. A confounding aspect in creating a general technique based on co-suppression is that some trans genes in plants lead to suppression of the endogenous locus and some do not. Results in C. elegans and Drosophila indicate that

60 for genetic inhibition. RNA containing a nucleotide sequences identical to a portion of the target gene is preferred for inhibition. RNA sequences with insertions, deletions, and single point mutations relative to the target sequence have also been found to be effective for inhibition. Thus, sequence

65 identity may optimized by alignment algorithms known in the art and calculating the percent difference between the nucle­otide sequences. Alternatively, the duplex region of the RNA

Page 16: 112   andrew fire - 7622633 - genetic inhibition by double-stranded rna

US 7,622,633 B2 5

may be defined functionally as a nucleotide sequence that is capable of hybridizing with a portion of the target gene tran­script.

The cell with the target gene may be derived from or contained in any organism (e.g., plant, animal, protozoan, virus, bacterium, or fungus). RNA may be synthesized either in vivo or in vitro. Endogenous RNA polymerase of the cell may mediate transcription in vivo, or cloned RNA poly­merase can be used for transcription in vivo or in vitro. For transcription from a transgene in vivo or an expression con- 10

struct, a regulatory region may be used to transcribe the RNA strand (or strands).

The RNA may be directly introduced into the cell (i.e., intracellularly); or introduced extracellularly into a cavity, interstitial space, into the circulation of an organism, intra- 15

duced orally, or may be introduced by bathing an organism in a solution containing RNA. Methods for oral introduction include direct mixing of RNA with food of the organism, as well as engineered approaches in which a species that is used as food is engineered to express an RNA, then fed to the 20

organism to be affected. Physical methods of introducing nucleic acids include injection directly into the cell or extra­cellular injection into the organism of an RNA solution.

The advantages of the present invention include: the ease of introducing double-stranded RNA into cells, the low concen- 25

tration of RNA which can be used, the stability of double­stranded RNA, and the effectiveness of the inhibition. The ability to use a low concentration of a naturally-occurring nucleic acid avoids several disadvantages of anti-sense inter­ference. This invention is not limited to in vitro use or to 30

6 drial reporter construct. Panel H shows a typical adult, with nuclear GFP-LacZ lacking in almost all body-wall muscles but retained in vulval muscles. Scale bars are 20 f.tm.

FIGS. 3 A-D show effects of double-stranded RNA corre­sponding to mex-3 on levels of the endogenous mRNA. Micrographs show in situ hybridization to embryos (dark stain). Panel A: Negative control showing lack of staining in the absence of hybridization probe. Panel B: Embryo from uninjected parent (normal pattern of endogenous mex-3 RNA20

). Panel C: Embryo from a parent injected with puri­fied mex-3B antisense RNA. These embryos and the parent animals retain the mex-3 mRNA, although levels may have been somewhat less than wild type. Panel D: Embryo from a parent injected with dsRNA corresponding to mex-3B; no mex-3 RNA was detected. Scale: each embryo is approxi­mately 50 f.tm in length.

FIG. 4 shows inhibitory activity ofunc-22A as a function of structure and concentration. The main graph (A) indicates fractions in each behavioral class. Embryos in the uterus and already covered with an eggshell at the time of injection are not affected and, thus, are not included. Progeny cohort groups are labels 1 for0-6 hours, 2 for 6-15 hours, 3 for 15-27 hours, 4 for 27-41 hours, and 5 for 41-56 hours. The bottom­left diagram (B) shows genetically derived relationship between unc-22 gene dosage and behavior based on analyses ofunc-22 heterozygotes and polyploids8

•3

.

FIGS. 5 A-C show examples of genetic inhibition follow-ing ingestion by C. elegans of dsRNAs from expressing bac­teria. Panel A: General strategy for production of dsRNA by cloning a segment of interest between flanking copies of the bacteriophage T7 promoter and transcribing both strands of the segment by transfecting a bacterial strain (BL21/DE3)28

expressing the T7 polymerase gene from an inducible (Lac) promoter. Panel B: A GFP-expressing C. elegans strain,

specific sequence compositions, as are techniques based on triple-strand formation. And unlike antisense interference, triple-strand interference, and co-suppression, this invention does not suffer from being limited to a particular set of target genes, a particular portion of the target gene's nucleotide sequence, or a particular trans gene or viral delivery method. These concerns have been a serious obstacle to designing general strategies according to the prior art for inhibiting gene expression of a target gene of interest.

35 PD4251 (see FIG. 2), fed on a native bacterial host. Panel C: PD4251 animals reared on a diet of bacteria expressing dsRNA corresponding to the coding region for gfp.

DETAILED DESCRIPTION OF THE INVENTION Furthermore, genetic manipulation becomes possible in 40

organisms that are not classical genetic models. Breeding and screening programs may be accelerated by the ability to rap­idly assay the consequences of a specific, targeted gene dis­ruption. Gene disruptions may be used to discover the func­tion of the target gene, to produce disease models in which the 45

target gene are involved in causing or preventing a pathologi-

The present invention provides a method of producing sequence-specific inhibition of gene expression by introduc­ing double-stranded RNA ( dsRNA). A process is provided for inhibiting expression of a target gene in a cell. The process comprises introduction of RNA with partial or fully double­stranded character into the cell. Inhibition is sequence-spe-

cal condition, and to produce organisms with improved eco­nomic properties.

BRIEF DESCRIPTION OF THE DRAWINGS

FIG. 1 shows the genes used to study RNA-mediated genetic inhibition in C. elegans. Intron-exon structure for genes used to test RNA-mediated inhibition are shown (ex­ons: filled boxes; introns: open boxes; 5' and 3' untranslated regions: shaded; unc-229

, unc-54 12, fem-1 14

, and hlh-1 15).

FIGS. 2 A-I show analysis of inhibitory RNA effects in individual cells. These experiments were carried out in a reporter strain (called PD4251) expressing two different reporter proteins, nuclear GFP-LacZ and mitochondrial GFP. The micrographs show progeny of injected animals visual­ized by a fluorescence microscope. Panels A (young larva), B (adult), and C (adult body wall; high magnification) result from injection of a control RNA (ds-unc22A). Panels D-F show progeny of animals injected with ds-gfpG. Panels G-I demonstrate specificity. Animals are injected with ds-lacZL RNA, which should affect the nuclear but not the mitochon-

cific in that a nucleotide sequence from a portion of the target gene is chosen to produce inhibitory RNA. We disclose that this process is (1) effective in producing inhibition of gene

50 expression, (2) specific to the targeted gene, and (3) general in allowing inhibition of many different types of target gene.

The target gene may be a gene derived from the cell (i.e., a cellular gene), an endogenous gene (i.e., a cellular gene present in the genome), a trans gene (i.e., a gene construct

55 inserted at an ectopic site in the genome of the cell), or a gene from a pathogen which is capable of infecting an organism from which the cell is derived. Depending on the particular target gene and the dose of double stranded RNA material delivered, this process may provide partial or complete loss of

60 function for the target gene. A reduction or loss of gene expression in at least 99% of targeted cells has been shown.

Inhibition of gene expression refers to the absence (or observable decrease) in the level of protein and/or mRNA product from a target gene. Specificity refers to the ability to

65 inhibit the target gene without manifest effects on other genes of the cell. The consequences of inhibition can be confirmed by examination of the outward properties of the cell or organ-

Page 17: 112   andrew fire - 7622633 - genetic inhibition by double-stranded rna

US 7,622,633 B2 7 8

known in the art (see Gribskov and Devereux, Sequence Analysis Primer, Stockton Press, 1991, and references cited therein) and calculating the percent difference between the nucleotide sequences by, for example, the Smith-Waterman algorithm as implemented in the BESTFIT software program using default parameters (e.g., University of Wisconsin Genetic Computing Group). Greater than 90% sequence identity, or even 100% sequence identity, between the inhibi-tory RNA and the portion of the target gene is preferred. Alternatively, the duplex region of the RNA may be defined functionally as a nucleotide sequence that is capable of hybridizing with a portion of the target gene transcript (e.g., 400 mM NaCl, 40 mM PIPES pH 6.4, 1 mM EDTA, 50° C. or 70° C. hybridization for 12-16 hours; followed by washing). The length of the identical nucleotide sequences may be at least 25, 50, 100, 200, 300 or 400 bases.

ism (as presented below in the examples) or by biochemical techniques such as RNA solution hybridization, nuclease pro­tection, Northern hybridization, reverse transcription, gene expression monitoring with a microarray, antibody binding, enzyme linked immunosorbent assay (ELISA), Western blot­ting, radioimmunoassay (RIA), other immunoassays, and fluorescence activated cell analysis (FACS). For RNA-medi­ated inhibition in a cell line or whole organism, gene expres­sion is conveniently assayed by use of a reporter or drug resistance gene whose protein product is easily assayed. Such 10

reporter genes include acetohydroxyacid synthase (AHAS), alkaline phosphatase (AP), beta galactosidase (LacZ), beta glucoronidase (GUS), chloramphenicol acetyltransferase (CAT), green fluorescent protein (GFP), horseradish peroxi­dase (HRP), luciferase (Luc ), nopaline synthase (NOS), 15

octopine synthase (OCS), and derivatives thereof. Multiple selectable markers are available that confer resistance to ampicillin, bleomycin, chloramphenicol, gentamycin, hygro­mycin, kanamycin, lincomycin, methotrexate, phosphino­thricin, puromycin, and tetracyclin.

As disclosed herein, 100% sequence identity between the RNA and the target gene is not required to practice the present invention. Thus the invention has the advantage of being able

20 to tolerate sequence variations that might be expected due to genetic mutation, strain polymorphism, or evolutionary divergence.

Depending on the assay, quantitation of the amount of gene expression allows one to determine a degree of inhibition which is greater than 10%, 33%, 50%, 90%, 95% or 99% as compared to a cell not treated according to the present inven­tion. Lower doses of injected material and longer times after administration of dsRNA may result in inhibition in a smaller fraction of cells (e.g., at least 10%, 20%, 50%, 7 5%, 90%, or 95% of targeted cells). Quantitation of gene expression in a cell may show similar amounts of inhibition at the level of accumulation of target mRNA or translation of target protein. As an example, the efficiency of inhibition may be deter­mined by assessing the amount of gene product in the cell: mRNA may be detected with a hybridization probe having a nucleotide sequence outside the region used for the inhibitory double-stranded RNA, or translated polypeptide may be detected with an antibody raised against the polypeptide sequence of that region.

The RNA may comprise one or more strands of polymer­ized ribonucleotide. It may include modifications to either the phosphate-sugar backbone or the nucleoside. For example, the phosphodiester linkages of natural RNA may be modified to include at least one of a nitrogen or sulfur heteroatom. Modifications in RNA structure may be tailored to allow specific genetic inhibition while avoiding a general panic response in some organisms which is generated by dsRNA. Likewise, bases may be modified to block the activity of adenosine deaminase. RNA may be produced enzymatically or by partial/total organic synthesis, any modified ribonucle­otide can be introduced by in vitro enzymatic or organic synthesis.

The double-stranded structure may be formed by a single self-complementary RNA strand or two complementary RNA strands. RNA duplex formation may be initiated either inside or outside the cell. The RNA may be introduced in an amount which allows delivery of at least one copy per cell. Higher doses (e.g., at least 5, 10, 100, 500 or 1000 copies per cell) of double-stranded material may yield more effective inhibition; lower doses may also be useful for specific appli­cations. Inhibition is sequence-specific in that nucleotide sequences corresponding to the duplex region of the RNA are targeted for genetic inhibition.

RNA containing a nucleotide sequences identical to a por­tion of the target gene are preferred for inhibition. RNA sequences with insertions, deletions, and single point muta­tions relative to the target sequence have also been found to be effective for inhibition. Thus, sequence identity may opti­mized by sequence comparison and alignment algorithms

The cell with the target gene may be derived from or contained in any organism. The organism may a plant, ani-

25 mal, protozoan, bacterium, virus, or fungus. The plant maybe a monocot, dicot or gynmosperm; the animal may be a ver­tebrate or invertebrate. Preferred microbes are those used in agriculture or by industry, and those that are pathogenic for plants or animals. Fungi include organisms in both the mold

30 and yeast morphologies. Plants include arabidopsis; field crops (e.g., alfalfa, barley,

bean, com, cotton, flax, pea, rape, rice, rye, safflower, sor­ghum, soybean, sunflower, tobacco, and wheat); vegetable crops (e.g., asparagus, beet, broccoli, cabbage, carrot, cauli-

35 flower, celery, cucumber, eggplant, lettuce, onion, pepper, potato, pumpkin, radish, spinach, squash, taro, tomato, and zucchini); fruit and nut crops (e.g., almond, apple, apricot, banana, blackberry, blueberry, cacao, cherry, coconut, cran­berry, date, fajoa, filbert, grape, grapefruit, guava, kiwi,

40 lemon, lime, mango, melon, nectarine, orange, papaya, pas­sion fruit, peach, peanut, pear, pineapple, pistachio, plum, raspberry, strawberry, tangerine, walnut, and watermelon); and ornamentals (e.g., alder, ash, aspen, azalea, birch, box­wood, camellia, carnation, chrysanthemum, elm, fir, ivy, jas-

45 mine, juniper, oak, palm, poplar, pine, redwood, rhododen­dron, rose, and rubber).

Examples of vertebrate animals include fish, mammal, cattle, goat, pig, sheep, rodent, hamster, mouse, rat, primate, and human; invertebrate animals include nematodes, other

so worms, drosophila, and other insects. Representative generae of nematodes include those that infect animals (e.g., Ancy­lostoma, Ascaridia, Ascaris, Bunostomum, Caenorhabditis, Capillaria, Chabertia, Cooperia, Dictyocaulus, Haemon­chus, Heterakis, Nematodirus, Oesophagostomum, Osterta-

55 gia, Oxyuris, Parascaris, Strongylus, Toxascaris, Trichuris, Trichostrongylus, Tjhchonema, Toxocara, Uncinaria) and those that infect plants (e.g., Bursaphalenchus, Cricone­mella, Diiylenchus, Ditylenchus, Globodera, Helicotylen­chus, Heterodera, Longidorus, Melodoigyne, Nacobbus,

60 Paratylenchus, Pratylenchus, Radopholus, Rotelynchus, Tylenchus, andXiphinema). Representative orders of insects include Coleoptera, Diptera, Lepidoptera, and Homoptera.

The cell having the target gene may be from the germ line or somatic, totipotent or pluripotent, dividing or non-divid-

65 ing, parenchyma or epithelium, immortalized or transformed, or the like. The cell may be a stem cell or a differentiated cell. Cell types that are differentiated include adipocytes, fibro-

Page 18: 112   andrew fire - 7622633 - genetic inhibition by double-stranded rna

US 7,622,633 B2 9

blasts, myocytes, cardiomyocytes, endothelium, neurons, glia, blood cells, megakaryocytes, lymphocytes, macroph­ages, neutrophils, eosinophils, basophils, mast cells, leuko­cytes, granulocytes, keratinocytes, chondrocytes, osteoblasts, osteoclasts, hepatocytes, and cells of the endocrine or exo­crine glands.

RNA may be synthesized either in vivo or in vitro. Endog­enous RNA polymerase of the cell may mediate transcription

10 encoded by the expression construct. Other methods known in the art for introducing nucleic acids to cells may be used, such as lipid-mediated carrier transport, chemical-mediated transport, such as calcium phosphate, and the like. Thus the RNA may be introduced along with components that perform one or more of the following activities: enhance RNA uptake by the cell, promote annealing of the duplex strands, stabilize the annealed strands, or other-wise increase inhibition of the

in vivo, or cloned RNA polymerase can be used for transcrip­tion in vivo or in vitro. For transcription from a transgene in 10

vivo or an expression construct, a regulatory region (e.g., promoter, enhancer, silencer, splice donor and acceptor, poly­adenylation) may be used to transcribe the RNA strand (or strands). Inhibition may be targeted by specific transcription

target gene. The present invention may be used to introduce RNA into

a cell for the treatment or prevention of disease. For example, dsRNA may be introduced into a cancerous cell or tumor and thereby inhibit gene expression of a gene required for main­tenance of the carcinogenic/tumorigenic phenotype. To pre-

in an organ, tissue, or cell type; stimulation of an environ­mental condition (e.g., infection, stress, temperature, chemi­cal inducers); and/or engineering transcription at a develop­mental stage or age. The RNA strands may or may not be polyadenylated; the RNA strands may or may not be capable

15 vent a disease or other pathology, a target gene may be selected which is required for initiation or maintenance of the disease/pathology. Treatment would include amelioration of any symptom associated with the disease or clinical indica-tion associated with the pathology.

of being translated into a polypeptide by a cell's translational 20

apparatus. RNA may be chemically or enzymatically synthe­sized by manual or automated reactions. The RNA may be synthesized by a cellular RNA polymerase or a bacteriophage RNA polymerase (e.g., T3, T7, SP6). The use and production

A gene derived from any pathogen may be targeted for inhibition. For example, the gene could cause immunosup­pression of the host directly or be essential for replication of the pathogen, transmission of the pathogen, or maintenance of the infection. The inhibitory RNA could be introduced in

of an expression construct are known in the art32•

33•

34 (see also WO 97/32016; U.S. Pat. Nos. 5,593,874, 5,698,425, 5,712,135, 5,789,214, and5,804,693; and the references cited therein). If synthesized chemically or by in vitro enzymatic synthesis, the RNA may be purified prior to introduction into the cell. For example, RNA can be purified from a mixture by extraction with a solvent or resin, precipitation, electrophore­sis, chromatography, or a combination thereof. Alternatively, the RNA may be used with no or a minimum of purification to avoid losses due to sample processing. The RNA may be dried for storage or dissolved in an aqueous solution. The solution may contain buffers or salts to promote annealing, and/or stabilization of the duplex strands.

25 cells in vitro or ex vivo and then subsequently placed into an animal to affect therapy, or directly treated by in vivo admin­istration. A method of gene therapy can be envisioned. For example, cells at risk for infection by a pathogen or already infected cells, particularly human immunodeficiency virus

30 (HIV) infections, may be targeted for treatment by introduc­tion ofRNA according to the invention. The target gene might be a pathogen or host gene responsible for entry of a pathogen into its host, drug metabolism by the pathogen or host, repli­cation or integration of the pathogen's genome, establishment

35 or spread of an infection in the host, or assembly of the next generation of pathogen. Methods of prophylaxis (i.e., preven­tion or decreased risk of infection), as well as reduction in the frequency or severity of symptoms associated with infection, can be envisioned.

RNA may be directly introduced into the cell (i.e., intrac­ellularly); or introduced extracellularly into a cavity, intersti­tial space, into the circulation of an organism, introduced 40

orally, or may be introduced by bathing an organism in a solution containing the RNA. Methods for oral introduction include direct mixing of the RNA with food of the organism,

The present invention could be used for treatment or devel-opment of treatments for cancers of any type, including solid tumors and leukemias, including: apudoma, choristoma, branchioma, malignant carcinoid syndrome, carcinoid heart disease, carcinoma (e.g., Walker, basal cell, basosquamous, as well as engineered approaches in which a species that is

used as food is engineered to express the RNA, then fed to the organism to be affected. For example, the RNA may be sprayed onto a plant or a plant may be genetically engineered to express the RNA in an amount sufficient to kill some or all of a pathogen known to infect the plant. Physical methods of introducing nucleic acids, for example, injection directly into the cell or extracellular injection into the organism, may also be used. We disclose herein that in C. elegans, double­stranded RNA introduced outside the cell inhibits gene expression. Vascular or extravascular circulation, the blood or lymph system, the phloem, the roots, and the cerebrospinal fluid are sites where the RNA may be introduced. A trans­genic organism that expresses RNA from a recombinant con­struct may be produced by introducing the construct into a zygote, an embryonic stem cell, or another multipotent cell derived from the appropriate organism.

Physical methods of introducing nucleic acids include injection of a solution containing the RNA, bombardment by particles covered by the RNA, soaking the cell or organism in a solution of the RNA, or electroporation of cell membranes in the presence of the RNA. A viral construct packaged into a viral particle would accomplish both efficient introduction of an expression construct into the cell and transcription of RNA

45 Brown-Pearce, ductal, Ehrlich tumor, in situ, Krebs 2, Merkel cell, mucinous, non-small cell lung, oat cell, papillary, scir­rhous, bronchiolar, bronchogenic, squamous cell, and transi­tional cell), histiocytic disorders, leukemia (e.g., B cell, mixed cell, null cell, T cell, T-ee!! chronic, HTLV-II-associ-

50 ated, lymphocytic acute, lymphocytic chronic, mast cell, and myeloid), histiocytosis malignant, Hodgkin disease, immu­noproliferative small, non-Hodgkin lymphoma, plasmacy­toma, reticuloendotheliosis, melanoma, chondroblastoma, chondroma, chondrosarcoma, fibroma, fibrosarcoma, giant

55 cell tumors, histiocytoma, lipoma, liposarcoma, mesothe­lioma, myxoma, myxosarcoma, osteoma, osteosarcoma, Ewing sarcoma, synovioma, adenofibroma, adenolym­phoma, carcinosarcoma, chordoma, cranio-pharyngioma, dysgerminoma, hamartoma, mesenchymoma, mesoneph-

60 roma, myosarcoma, ameloblastoma, cementoma, odontoma, teratoma, thymoma, trophoblastic tumor, adenocarcinoma, adenoma, cholangioma, cholesteatoma, cylindroma, cystad­enocarcinoma, cystadenoma, granulosa cell tumor, gynan­droblastoma, hepatoma, hidradenoma, islet cell tumor, Ley-

65 dig cell tumor, papilloma, Sertoli cell tumor, theca cell tumor, leiomyoma, leiomyosarcoma, myoblastoma, myoma, myo­sarcoma, rhabdomyoma, rhabdomyosarcoma, sarcoma,

Page 19: 112   andrew fire - 7622633 - genetic inhibition by double-stranded rna

US 7,622,633 B2 11

ependymoma, ganglioneuroma, glioma, medulloblastoma, meningioma, neurilemmoma, neuroblastoma, neuroepithe­lioma, neurofibroma, neuroma, paraganglioma, paragan­glioma nonchromaffin, angiokeratoma, angiolymphoid hyperplasia with eosinophilia, angioma sclerosing, angioma­tosis, glomangioma, hemangioendothelioma, hemangioma, hemangiopericytoma, hemangiosarcoma, lymphangioma, lymphangiomyoma, lymphangiosarcoma, pinealoma, carci­nosarcoma, chondrosarcoma, cystosarcoma phyllodes, fibro­sarcoma, hemangiosarcoma, leiomyosarcoma, leukosar­coma, liposarcoma, lymphangiosarcoma, myosarcoma, myxosarcoma, ovarian carcinoma, rhabdomyosarcoma, sar­coma (e.g., Ewing, experimental, Kaposi, and mast cell), neoplasms (e.g., bone, breast, digestive system, colorectal, liver, pancreatic, pituitary, testicular, orbital, head and neck, central nervous system, acoustic, pelvic, respiratory tract, and urogenital), neurofibromatosis, and cervical dysplasia, and for treatment of other conditions in which cells have become immortalized or transformed. The invention could be used in

12 be magnified by targeting multiple different components. Metabolism may also be manipulated by inhibiting feedback control in the pathway or production of unwanted metabolic byproducts.

The present invention may be used to reduce crop destruc­tion by other plant pathogens such as arachnids, insects, nematodes, protozoans, bacteria, or fungi. Some such plants and their pathogens are listed in Index of Plant Diseases in the United States (U.S. Dept. of Agriculture Handbook No. 165,

10 1960); Distribution of Plant-Parasitic Nematode Species in North America (Society ofNematologists, 1985); and Fungi on Plants and Plant Products in the United States (American Phytopathological Society, 1989). Insects with reduced abil­ity to damage crops or improved ability to prevent other

15 destructive insects from damaging crops may be produced. Furthermore, some nematodes are vectors of plant pathogens, and may be attacked by other beneficial nematodes which have no effect on plants. Inhibition of target gene activity

combination with other treatment modalities, such as chemo- 20

therapy, cryotherapy, hyperthermia, radiation therapy, and the like.

could be used to delay or prevent entry into a particular developmental step (e.g., metamorphosis), if plant disease was associated with a particular stage of the pathogen's life cycle. Interactions between pathogens may also be modified by the invention to limit crop damage. For example, the ability of beneficial nematodes to attack their harmful prey

As disclosed herein, the present invention may is not lim­ited to any type of target gene or nucleotide sequence. But the following classes of possible target genes are listed for illus­trative purposes: developmental genes (e.g., adhesion mol­ecules, cyclin kinase inhibitors, Wnt family members, Pax family members, Winged helix family members, Hox family members, cytokines/lymphokines and their receptors, growth/differentiation factors and their receptors, neurotrans­mitters and their receptors); oncogenes (e.g., ABLI, BCLI, BCL2, BCL6, CBFA2, CBL, CSFIR, ERBA, ERBB, EBRB2, ETSI, ETSI, ETV6, FGR, FOS, FYN, HCR, HRAS, JUN, KRAS, LCK, LYN, MDM2, MLL, MYB, MYC, MYCLl, MYCN, NRAS, PIMI, PML, RET, SRC, TALl, TCL3, and YES); tumor suppressor genes (e.g., APC, BRCAI, BRCA2, MADH4, MCC, NFI, NF2, RBI, TP53, and WTl); and enzymes (e.g., ACC synthases and oxidases, ACP desaturases and hydroxy lases, ADP-glucose pyropho­rylases, ATPases, alcohol dehydrogenases, amylases, amylo­glucosidases, catalases, cellulases, chalcone synthases, chiti­nases, cyclooxygenases, decarboxylases, dextrinases, DNA and RNA polymerases, galactosidases, glucanases, glucose oxidases, granule-bound starch synthases, GTPases, heli­cases, hemicellulases, integrases, inulinases, invertases, isomerases, kinases, lactases, lipases, lipoxygenases, lysozymes, nopaline synthases, octopine synthases, pectinesterases, peroxidases, phosphatases, phospholipases, phosphorylases, phytases, plant growth regulator synthases, polygalacturonases, proteinases and peptidases, pullanases, recombinases, reverse transcriptases, RUBISCOs, topoi­somerases, and xylanases).

The present invention could comprise a method for pro­ducing plants with reduced susceptibility to climatic injury, susceptibility to insect damage, susceptibility to infection by a pathogen, or altered fruit ripening characteristics. The tar­geted gene may be an enzyme, a plant structural protein, a gene involved in pathogenesis, or an enzyme that is involved in the production of a non-proteinaceous part of the plant (i.e.,

25 may be enhanced by inhibition ofbehavior-controlling nema­tode genes according to the invention.

Although pathogens cause disease, some of the microbes interact with their plant host in a beneficial mam1er. For example, some bacteria are involved in symbiotic relation-

30 ships that fix nitrogen and some fungi produce phytohor­mones. Such beneficial interactions may be promoted by using the present invention to inhibit target gene activity in the plant and/or the microbe.

Another utility of the present invention could be a method 35 of identifYing gene function in an organism comprising the

use of double-stranded RNA to inhibit the activity of a target gene of previously unknown function. Instead of the time consuming and laborious isolation of mutants by traditional genetic screening, functional genomics would envision deter-

40 mining the function ofuncharacterized genes by employing the invention to reduce the amount and/or alter the timing of target gene activity. The invention could be used in determin­ing potential targets for pharmaceutics, understanding nor­mal and pathological events associated with development,

45 determining signaling pathways responsible for postnatal development/aging, and the like. The increasing speed of acquiring nucleotide sequence information from genomic and expressed gene sources, including total sequences for the yeast, D. melanogaster, and C. elegans genomes, can be

50 coupled with the invention to determine gene function in an organism (e.g., nematode). The preference of different organ­isms to use particular codons, searching sequence databases for related gene products, correlating the linkage map of genetic traits with the physical map from which the nude-

55 otide sequences are derived, and artificial intelligence meth­ods may be used to define putative open reading frames from the nucleotide sequences acquired in such sequencing projects.

A simple assay would be to inhibit gene expression accord-60 ing to the partial sequence available from an expressed

sequence tag (EST). Functional alterations in growth, devel­opment, metabolism, disease resistance, or other biological processes would be indicative of the normal role of the EST's

a carbohydrate or lipid). If an expression construct is used to transcribe the RNA in a plant, transcription by a wound- or stress-inducible; tissue-specific (e.g., fruit, seed, anther, flower, leaf, root); or otherwise regulatable (e.g., infection, light, temperature, chemical) promoter may be used. By inhibiting enzymes at one or more points in a metabolic 65

pathway or genes involved in pathogenesis, the effect may be enhanced: each activity will be affected and the effects may

gene product. The ease with which RNA can be introduced into an intact

cell/organism containing the target gene allows the present invention to be used in high throughput screening (HTS). For

Page 20: 112   andrew fire - 7622633 - genetic inhibition by double-stranded rna

US 7,622,633 B2 13

example, duplex RNA can be produced by an amplification reaction using primers flanking the inserts of any gene library derived from the target cell/organism. Inserts may be derived from genomic DNA or mRNA (e.g., eDNA and cRNA). Indi­vidual clones from the library can be replicated and then isolated in separate reactions, but preferably the library is maintained in individual reaction vessels (e.g., a 96-well microtiter plate) to minimize the number of steps required to practice the invention and to allow automation of the process. Solutions containing duplex RNAs that are capable of inhib- 10

iting the different expressed genes can be placed into indi­vidual wells positioned on a microtiter plate as an ordered array, and intact cells/organisms in each well can be-assayed for any changes or modifications in behavior or development due to inhibition of target gene activity. The amplified RNA 15

can be fed directly to, injected into, the cell/organism con­taining the target gene. Alternatively, the duplex RNA can be produced by in vivo or in vitro transcription from an expres­sion construct used to produce the library. The construct can be replicated as individual clones of the library and tran- 20

scribed to produce the RNA; each clone can then be fed to, or injected into, the cell/organism containing the target gene. The function of the target gene can be assayed from the effects it has on the cell/organism when gene activity is inhibited. This screening could be amenable to small subjects that can 25

be processed in large number, for example: arabidopsis, bac­teria, drosophila, fungi, nematodes, viruses, zebrafish, and tissue culture cells derived from mammals.

14 may be expressed in both membrane bound and secreted forms. Instead of isolating a nonsense mutation that termi­nates translation before the transmembrane domain, the func­tional consequences ofhaving only secreted hormone can be determined according to the invention by targeting the exon containing the transmembrane domain and thereby inhibiting expression of membrane-bound hormone.

The present invention may be used alone or as a component of a kit having at least one of the reagents necessary to carry out the in vitro or in vivo introduction of RNA to test samples or subjects. Preferred components are the dsRNA and a vehicle that promotes introduction of the dsRNA. Such a kit may also include instructions to allow a user of the kit to practice the invention.

Pesticides may include the RNA molecule itself, an expres­sion construct capable of expressing the RNA, or organisms transfected with the expression construct. The pesticide of the present invention may serve as an arachnicide, insecticide, nematicide, viricide, bactericide, and/or fungicide. For example, plant parts that are accessible above ground (e.g., flowers, fruits, buds, leaves, seeds, shoots, bark, stems) may be sprayed with pesticide, the soil may be soaked with pesti­cide to access plant parts growing beneath ground level, or the pest may be contacted with pesticide directly. If pests interact with each other, the RNA may be transmitted between them. Alternatively, if inhibition of the target gene results in a ben­eficial effect on plant growth or development, the aforemen­tioned RNA, expression construct, or transfected organism A nematode or other organism that produces a colorimet­

ric, fluorogenic, or luminescent signal in response to a regu­lated promoter (e.g., transfected with a reporter gene con­struct) can be assayed in an HTS format to identify DNA­binding proteins that regulate the promoter. In the assay's simplest form, inhibition of a negative regulator results in an increase of the signal and inhibition of a positive regulator results in a decrease of the signal.

30 may be considered a nutritional agent. In either case, genetic engineering of the plant is not required to achieve the objec­tives of the invention.

If a characteristic of an organism is determined to be genetically linked to a polymorphism through RFLP or QTL analysis, the present invention can be used to gain insight regarding whether that genetic polymorphism might be directly responsible for the characteristic. For example, a fragment defining the genetic polymorphism or sequences in the vicinity of such a genetic polymorphism can be amplified to produce an RNA, the duplex RNA can be introduced to the organism, and whether an alteration in the characteristic is correlated with inhibition can be determined. Of course, there may be trivial explanations for negative results with this type of assay, for example: inhibition of the target gene causes lethality, inhibition of the target gene may not result in any observable alteration, the fragment contains nucleotide sequences that are not capable of inhibiting the target gene, or the target gene's activity is redundant.

The present invention may be useful in allowing the inhi­bition of essential genes. Such genes may be required for cell or organism viability at only particular stages of development or cellular compartments. The functional equivalent of con­ditional mutations may be produced by inhibiting activity of the target gene when or where it is not required for viability. The invention allows addition of RNA at specific times of development and locations in the organism without introduc­ing permanent mutations into the target genome.

Alternatively, an organism may be engineered to produce dsRNA which produces commercially or medically benefi-

35 cia! results, for example, resistance to a pathogen or its patho­genic effects, improved growth, or novel developmental pat­terns.

Used as either an pesticide or nutrient, a formulation of the present invention may be delivered to the end user in dry or

40 liquid form: for example, as a dust, granulate, emulsion, paste, solution, concentrate, suspension, or encapsulation. Instructions for safe and effective use may also be provided with the formulation. The formulation might be used directly, but concentrates would require dilution by mixing with an

45 extender provided by the formulator or the end user. Simi­larly, an emulsion, paste, or suspension may require the end user to perform certain preparation steps before application. The formulation may include a combination of chemical additives known in the art such as solid carriers, minerals,

50 solvents, dispersants, surfactants, emulsifiers, tackifiers, binders, and other adjuvants. Preservatives and stabilizers may also be added to the formulation to facilitate storage. The crop area or plant may also be treated simultaneously or separately with other pesticides or fertilizers. Methods of

55 application include dusting, scattering or pouring, soaking, spraying, atomizing, and coating. The precise physical form and chemical composition of the formulation, and its method of application, would be chosen to promote the objectives of the invention and in accordance with prevailing circum-

60 stances. Expression constructs and transfected hosts capable of replication may also promote the persistence and/ or spread of the formulation. If alternative splicing produced a family of transcripts that

were distinguished by usage of characteristic exons, the present invention can target inhibition through the appropri­ate exons to specifically inhibit or to distinguish among the 65

functions of family members. For example, a hormone that contained an alternatively spliced transmembrane domain

Description of the dsRNA Inhibition Phenomenon in C. elegans

The operation of the present invention was shown in the model genetic organism Caenorhabditis elegans.

Page 21: 112   andrew fire - 7622633 - genetic inhibition by double-stranded rna

US 7,622,633 B2 15

Introduction of RNA into cells had been seen in certain biological systems to interfere with function of an endog­enous gene1

'2

, Many such effects were believed to result from a simple antisense mechanism dependent on hybridization between injected single-stranded RNA and endogenous tran­scripts, In other cases, a more complex mechanism had been suggested, One instance of an RNA-mediated mechanism was RNA interference (RNAi) phenomenon in the nematode C elegans, RNAi had been used in a variety of studies to manipulate gene expression3

'4

,

Despite the usefulness of RNAi in C elegans, many fea­tures had been difficult to explain, Also, the lack of a clear understanding of the critical requirements for interfering RNA led to a sporadic record of failure and partial success in attempts to extend RNAi beyond the earliest stages following injection, A statement frequently made in the literature was that sense and antisense RNA preparations are each sufficient

16 Purified antisense and sense RNAs covering a 742 nt seg­

ment ofunc-22 had only marginal inhibitory activity, requir­ing a very high dose of injected RNA for any observable effect (FIG, 4), By contrast, a sense+antisense mixture produced a highly effective inhibition of endogenous gene activity (FIG, 4), The mixture was at least two orders of magnitude more effective than either single strand in inhibiting gene expres­sion, The lowest dose of the sense+antisense mixture tested, approximately 60,000 molecules of each strand per adult, led

10 to twitching phenotypes in an average of 100 progeny, unc-22 expression begins in embryos with approximately 500 cells, At this point, the original injected material would be diluted to at most a few molecules per celL

The potent inhibitory activity of the sense+antisense mix-15 ture could reflect formation of double-stranded RNA

to cause interference3'4

, The only precedent for such a situa­tion was in plants where the process of co-suppression had a 20 similar history of usefulness in certain cases, failure in others, and no ability to design interference protocols with a high chance of success, Working with C elegans, we discovered

( dsRNA), or conceivably some alternate synergy between the strands, Electrophoretic analysis indicated that the injected material was predominantly double stranded, The dsRNA was gel purified from the annealed mixture and found to retain potent inhibitory activity, Although annealing prior to injection was compatible with inhibition, it was not neces-sary, Mixing of sense and antisense RNAs in low salt (under conditions of minimal dsRNA formation), or rapid sequential injection of sense and antisense strands, were sufficient to allow complete inhibition, A long interval(> 1 hour) between sequential injections of sense and antisense RNA resulted in a dramatic decrease in inhibitory activity, This suggests that injected single strands may be degraded or otherwise ren­dered inaccessible in the absence of the complementary

an RNA structure that would give effective and uniform genetic inhibition, The prior art did not teach or suggest that 25 RNA structure was a critical feature for inhibition of gene expression, Indeed the ability of crude sense and antisense preparations to produce interference3

'4 had been taken as an

indication that RNA structure was not a critical factor, Instead, the extensive plant literature and much of the ongo­ing research in C elegans was focused on the possibility that detailed features of the target gene sequence or its chromo­somallocale was the critical feature for interfering with gene expression,

30 strand, An issue of specificity arises when considering known

cellular responses to dsRNA, Some organisms have a dsRNA-dependent protein kinase that activates a panic response mechanism 10

, Conceivably, the inventive sense+ The inventors carefully purified sense or antisense RNA for

unc-22 and tested each for gene-specific inhibition, While the crude sense and antisense preparations had strong interfering activity, it was found that the purified sense and antisense RNAs had only marginal inhibitory activity, This was unex­pected because many techniques in molecular biology are based on the assumption that RNA produced with specific in vitro promoters ( e,g,, T3 or T7 RNA polymerase), or with characterized promoters in vivo, is produced predominantly from a single strand, The inventors had carried out purifica­tion of these crude preparations to investigate whether a small fraction of the RNA had an unusual structure which might be responsible for the observed genetic inhibition, To rigorously test whether double-stranded character might contribute to genetic inhibition, the inventors carried out additional purifi­cation of single-stranded RNAs and compared inhibitory activities of individual strands with that of the double­stranded hybrid,

The following examples are meant to be illustrative of the present invention; however, the practice of the invention is not limited or restricted in any way by them,

Analysis of RNA-Mediated Inhibition of C elegans Genes

The unc-22 gene was chosen for initial comparisons of activity as a result of previous genetic analysis that yields a semi-quantitative comparison between unc-22 gene activity and the movement phenotypes of animals3

'8

: decreases in activity produce an increasingly severe twitching phenotype, while complete loss of function results in the additional appearance of muscle structural defects and impaired motil­ity, unc-22 encodes an abundant but non-essential myofila­ment protein 7 -

9, unc-22 mRNA is present at several thousand

copies per striated muscle cell3,

35 antisense synergy could reflect a non-specific potentiation of antisense effects by such a panic mechanism, This was not found to be the case: co-injection of dsRNA segments unre­lated to unc-22 did not potentiate the ability ofunc-22 single strands to mediate inhibition, Also investigated was whether

40 double-stranded structure could potentiate inhibitory activity when placed in cis to a single-stranded segment No such potentiation was seen; unrelated double-stranded sequences located 5' or 3' of a single-stranded unc-22 segment did not stimulate inhibition, Thus potentiation of gene-specific inhi-

45 bition was observed only when dsRNA sequences exist within the region of homology with the target gene,

The phenotype produced by unc-22 dsRNA was specific, Progeny of injected animals exhibited behavior indistin­guishable from characteristic unc-22 loss of function

50 mutants, Target-specificity of dsRNA effects using three additional genes with well characterized phenotypes (FIG, 1 and Table 1), unc-54 encodes a body wall muscle myosin heavy chain isoform required for full muscle contraction 7

'11

'

12, fem-1 encodes an ankyrin-repeat containing protein 55 required in hermaphrodites for sperm production13

'14

, and hlh-1 encodes a C elegans homolog of the myoD family required for proper body shape and motility15

'16

, For each of these genes, injection of dsRNA produced progeny broods exhibiting the known null mutant phenotype, while the puri-

60 fied single strands produced no significant reduction in gene expression, With one exception, all of the phenotypic conse­quences of dsRNA injection were those expected from inhi­bition of the corresponding gene, The exception (segment unc54C, which led to an embryonic and larval arrest pheno-

65 type not seen with unc-54 null mutants) was illustrative, This segment covers the highly conserved myosin motor domain, and might have been expected to inhibit the activity of other

Page 22: 112   andrew fire - 7622633 - genetic inhibition by double-stranded rna

US 7,622,633 B2 17

highly related myosin heavy chain genes 17. This interpreta­

tion would support uses of the present invention in which nucleotide sequence comparison of dsRNA and target gene show less than 100% identity. The unc54C segment has been unique in our overall experience to date: effects of 18 other dsRNA segments have all been limited to those expected from characterized null mutants.

18 cavity or gonad of young adults also produced gene-specific inhibition in somatic tissues of the injected animal (Table 2).

Table 3 shows that C. elegans can respond in a gene­specific manner to dsRNA encountered in the environment. Bacteria are a natural food source for C. elegans. The bacteria are ingested, ground in the animal's pharynx, and the bacte­rial contents taken up in the gut. The results show that E. coli bacteria expressing dsRNAs can confer specific inhibitory effects on C. elegans nematode larvae that feed on them.

The strong phenotypes seen following dsRNA injection are indicative of inhibitory effects occurring in a high fraction of cells. The unc-54 and hlh-1 muscle phenotypes, in particular, 10

are known to result from a large number of defective muscle cells 11

•16

. To examine inhibitory effects of dsRNA on a cel­lular level, a transgenic line expressing two different GFP­derived fluorescent reporter proteins in body muscle was used. Injection of dsRNA directed to gfp produced dramatic decreases in the fraction of fluorescent cells (FIG. 2). Both reporter proteins were absent from the negative cells, while the few positive cells generally expressed both GFP forms.

Three C. elegans genes were analyzed. For each gene, corresponding dsRNA was expressed in E. coli by inserting a segment of the coding region into a plasmid construct designed for bidirectional transcription by bacteriophage T7 RNA polymerase. The dsRNA segments used for these

15 experiments were the same as those used in previous micro­injection experiments (see FIG.l). The effects resulting from feeding these bacteria to C. elegans were compared to the effects achieved by microinjecting animals with dsRNA.

The pattern of mosaicism observed with gfp inhibition was not random. At low doses of dsRNA, the inventors saw fre­quent inhibition in the embryonically-derived muscle cells present when the animal hatched. The inhibitory effect in these differentiated cells persisted through larval growth: these cells produced little or no additional GFP as the affected animals grew. The 14 postembryonically-derived striated muscles are born during early larval stages and were more resistant to inhibition. These cells have come through addi­tional divisions (13-14 versus 8-9 for embryonic muscles 18

"

19 ). At high concentrations of gfp dsRNA, inhibition was noted in virtually all striated bodywall muscles, with occa­sional single escaping cells including cells born in embryonic or post-embryonic stages. The nonstriated vulval muscles, born during late larval development, appeared resistant to genetic inhibition at all tested concentrations of injected RNA. The latter result is important for evaluating the use of the present invention in other systems. First, it indicates that failure in one set of cells from an organism does not neces­sarily indicate complete non-applicability of the invention to that organism. Second, it is important to realize that not all tissues in the organism need to be affected for the invention to be used in an organism. This may serve as an advantage in some situations.

A few observations serve to clarify the nature of possible targets and mechanisms for RNA-mediated genetic inhibition in C. elegans:

First, dsRNA segments corresponding to a variety of intron and promoter sequences did not produce detectable inhibition (Table 1). Although consistent with possible inhibition at a post-transcriptionallevel, these experiments do not rule out inhibition at the level of the gene.

Second, dsRNA injection produced a dramatic decrease in the level of the endogenous mRNA transcript (FIG. 3). Here, a mex-3 transcript that is abundant in the gonad and early embryos20 was targeted, where straightforward in situ hybrid­ization can beperformed5

. No endogenous mex-3 mRNA was observed in animals injected with a dsRNA segment derived from mex-3 (FIG. 3D), but injection of purified mex-3 anti­sense RNA resulted in animals that retained substantial endogenous mRNA levels (FIG. 3C).

Third, dsRNA-mediated inhibition showed a surprising ability to cross cellular boundaries. Injection of dsRNA for unc-22, gfp, or lacZ into the body cavity of the head or tail produced a specific and robust inhibition of gene expression in the progeny brood (Table 2). Inhibition was seen in the progeny of both gonad arms, ruling out a transient "nicking" of the gonad in these injections. dsRNA injected into body

20

25

30

35

The C. elegans gene unc-22 encodes an abundant muscle filament protein. unc-22 null mutations produce a character-istic and uniform twitching phenotype in which the animals can sustain only transient muscle contraction. When wild­type animals were fed bacteria expressing a dsRNA segment from unc-22, a high fraction (85%) exhibited a weak but still distinct twitching phenotype characteristic of partial loss of function for the unc-22 gene. The C. elegans fem-1 gene encodes a late component of the sex determination pathway. Null mutations prevent the production of sperm and lead euploid (XX) animals to develop as females, while wild type XX animals develop as hermaphrodites. When wild-type ani-mals were fed bacteria expressing dsRNA corresponding to fem-1, a fraction ( 43%) exhibit a sperm-less (female) pheno­type and were sterile. Finally, the ability to inhibit gene expression of a transgene target was assessed. When animals carrying a gfp transgene were fed bacteria expressing dsRNA corresponding to the gfp reporter, an obvious decrease in the overall level of GFP fluorescence was observed, again in approximately 12% of the population (see FIG. 5, panels B

40 and C).

The effects of these ingested RNAs were specific. Bacteria carrying different dsRNAs from fem-1 and gfp produced no twitching, dsRNAs from unc-22 andfem-1 did not reduce gfp expression, and dsRNAs from gfp and unc-22 did not produce

45 females. These inhibitory effects were apparently mediated by dsRNA: bacteria expressing only the sense or antisense strand for either gfp or unc-22 caused no evident phenotypic effects on their C. elegans predators.

Table 4 shows the effects ofbathing C. elegans in a solution 50 containing dsRNA. Larvae were bathed for 24 hours in solu­

tions of the indicated dsRNAs (1 mg/ml), then allowed to recover in normal media and allowed to grow under standard conditions for two days. The unc-22 dsRNA was segment ds-unc22A from FIG. 1. pos-1 and sqt-3 dsRNAs were from

55 the full length eDNA clones. pos-1 encodes an essential maternally provided component required early in embyogen­esis. Mutations removing pos-1 activity have an early embry­onic arrest characteristic of skn-like mutations29

•30

. Cloning and activity patterns for sqt-3 have been described31

. C. 60 elegans sqt-3 mutants have mutations in the coi-l collagen

gene31. Phenotypes of affected animals are noted. Incidences

of clear phenotypic effects in these experiments were 5-l 0% for unc-22, 50% for pos-1, and 5% for sqt-3. These are fre­quencies of unambiguous phenocopies; other treated animals

65 may have had marginal defects corresponding to the target gene that were not observable. Each treatment was fully gene­specific in that unc-22 dsRNA produced only Unc-22 pheno-

Page 23: 112   andrew fire - 7622633 - genetic inhibition by double-stranded rna

US 7,622,633 B2 19

types, pos-1 dsRNA produced only Pos-1 phenotypes, and sqt-3 dsRNA produced only Sqt-3 phenotypes.

Some of the results described herein were published after the filing of our provisional application. Those publications and a review can be cited as Fire, A., et a!. Nature, 391, 806-811, 1998; Timmons, L. & Fire, A. Nature, 395, 854, 1998; and Montgomery, M. K. & Fire, A. Trends in Genetics, 14, 255-258, 1998.

The effects described herein significantly augment avail­able tools for studying gene function in C. elegans and other organisms. In particular, functional analysis should now be possible for a large number of interesting coding regions21 for which no specific function have been defined. Several of these observations show the properties of dsRNA that may affect the design of processes for inhibition of gene expression. For example, one case was observed in which a nucleotide sequence shared between several myosin genes may inhibit gene expression of several members of a related gene family.

Methods of RNA Synthesis and Microinjection RNA was synthesized from phagemid clones with T3 and

T7 RNA polymerase6, followed by template removal with

two sequential DNase treatments. In cases where sense, anti­sense, and mixed RNA populations were to be compared, RNAs were further purified by electrophoresis on low-gel­ling-temperature agarose. Gel-purified products appeared to lack many of the minor bands seen in the original "sense" and "antisense" preparations. Nonetheless, RNA species accounting for less than 10% of purified RNA preparations would not have been observed. Without gel purification, the "sense" and "antisense" preparations produced significant inhibition. This inhibitory activity was reduced or eliminated upon gel purification. By contrast, sense+antisense mixtures of gel purified and non-gel-purified RNA preparations pro­duced identical effects.

20 Methods for Analysis of Phenotypes

Inhibition of endogenous genes was generally assayed in a wild type genetic background (N2). Features analyzed included movement, feeding, hatching, body shape, sexual identity, and fertility. Inhibition with gfp27 and lacZ activity was assessed using strain PD4251. This strain is a stable transgenic strain containing an integrated array (ccls4251) made up of three plasmids: pSAK4 (myo-3 promoter driving mitochondrially targeted GFP), pSAK2 (myo-3 promoter

10 driving a nuclear targeted GFP-LacZ fusion), and a dpy-20 subclone26 as a selectable marker. This strain produces GFP in all body muscles, with a combination of mitochondrial and nuclear localization. The two distinct compartments are eas­ily distinguished in these cells, allowing a facile distinction

15 between cells expressing both, either, or neither of the origi­nal GFP constructs.

Gonadal injection was performed by inserting the micro­injection needle into the gonadal syncitium of adults and expelling 20-100 pi of solution (see Reference 25). Body

20 cavity injections followed a similar procedure, with needle insertion into regions of the head and tail beyond the positions of the two gonad arms. Injection into the cytoplasm of intes­tinal cells was another effective means of RNA delivery, and may be the least disruptive to the animal. After recovery and

25 transfer to standard solid media, injected animals were trans­ferred to fresh culture plates at 16 hour intervals. This yields a series of semi-synchronous cohorts in which it was straight­forward to identify phenotypic differences. A characteristic temporal pattern of phenotypic severity is observed among

30 progeny. First, there is a short "clearance" interval in which unaffected progeny are produced. These include imperme­able fertilized eggs present at the time of injection. After the clearance period, individuals are produced which show the inhibitory phenotype. After injected animals have produced

35 eggs for several days, gonads can in some cases "revert" to produce incompletely affected or phenotypically normal Following a short (5 minute) treatment at 68° C. to remove

secondary structure, sense+antisense annealing was carried out in injection buffer27 at 37° C. for 10-30 minutes. Forma­tion of predominantly double stranded material was con­firmed by testing migration on a standard (non-denaturing) 40 agarose gel: for each RNA pair, gel mobility was shifted to that expected for double-stranded RNA of the appropriate length. Co-incubation of the two strands in a low-salt buffer ( 5 mM Tris-HCI pH 7.5, 0.5 mM EDTA) was insufficient for visible formation of double-stranded RNA in vitro. Non- 45

progeny.

Additional Description of the Results FIG. 1 shows genes used to study RNA-mediated genetic

inhibition in C. elegans. Intron-exon structure for genes used to test RNA-mediated inhibition are shown (exons: filled boxes; introns: open boxes; 5' and 3' untranslated regions: shaded; sequence references are as follows: unc-229

, unc-5412, fem-1 14

, and hlh-1 15). These genes were chosen based

on: (1) a defined molecular structure, (2) classical genetic annealed sense+antisense RNAs for unc22B and gfpG were tested for inhibitory effect and found to be much more active than the individual single strands, but 2-4 fold less active than equivalent pre-annealed preparations.

After pre-annealing of the single strands for unc22A, the single electrophoretic species corresponding in size to that expected for dsRNA was purified using two rounds of gel electrophoresis. This material retained a high degree of inhibitory activity.

Except where noted, injection mixes were constructed so animals would receive an average of0.5x106 to l.Ox106 mol­ecules of RNA. For comparisons of sense, antisense, and dsRNA activities, injections were compared with equal masses of RNA (i.e., dsRNA at half the molar concentration of the single strands). Numbers of molecules injected per adult are given as rough approximations based on concentra­tion of RNA in the injected material (estimated from ethidium bromide staining) and injection volume (estimated from vis­ible displacement at the site of injection). A variability of several-fold in injection volume between individual animals is possible; however, such variability would not affect any of the conclusions drawn herein.

data showing the nature of the null phenotype. Each segment tested for inhibitory effects is designated with the name of the gene followed by a single letter (e.g., unc22C). Segments

50 derived from genomic DNA are shown above the gene, seg­ments derived from eDNA are shown below the gene. The consequences of injecting double-stranded RNA segments for each of these genes is described in Table 1. dsRNA sequences from the coding region of each gene produced a

55 phenotype resembling the null phenotype for that gene. The effects of inhibitory RNA were analyzed in individual

cells (FIG. 2, panels A-H). These experiments were carried out in a reporter strain (called PD4251) expressing two dif­ferent reporter proteins: nuclear GFP-LacZ and mitochon-

60 drial GFP, both expressed in body muscle. The fluorescent nature of these reporter proteins allowed us to examine indi­vidual cells under the fluorescence microscope to determine the extent and generality of the observed inhibition of gene. ds-unc22A RNA was injected as a negative control. GFP

65 expression in progeny of these injected animals was not affected. The GFP patterns of these progeny appeared iden­tical to the parent strain, with prominent fluorescence in

Page 24: 112   andrew fire - 7622633 - genetic inhibition by double-stranded rna

US 7,622,633 B2 21

nuclei (the nuclear localized GFP-LacZ) and mitochondria (the mitochondrially targeted GFP): young larva (FIG. 2A), adult (FIG. 2B), and adult body wall at high magnification (FIG. 2C).

In contrast, the progeny of animals injected with ds-gfpG 5

RNA are affected (FIGS. 2D-F). Observable GFP fluores­cence is completely absent in over 95% of the cells. Few active cells were seen in larvae (FIG. 2D shows a larva with one active cell; uninjected controls show GFP activity in all 81 body wall muscle cells). Inhibition was not effective in all 10

tissues: the entire vulval musculature expressed active GFP in an adult animal (FIG. 2E). Rare GFP positive body wall muscle cells were also seen adult animals (two active cells are shown in FIG. 2F). Inhibition was target specific (FIGS. 2G-I). Animals were injected with ds-lacZL RNA, which 15

should affect the nuclear but not the mitochondrial reporter construct. In the animals derived from this injection, mito­chondrial-targeted GFP appeared unaffected while the nuclear-targeted GFP-LacZ was absent from almost all cells (larva in FIG. 2G). A typical adult lacked nuclear GFP-LacZ 20

in almost all body-wall muscles but retained activity in vulval muscles (FIG. 2H). Scale bars in FIG. 2 are 20 fllll·

The effects of double-stranded RNA corresponding to mex-3 on levels of the endogenous mRNA was shown by in situ hybridization to embryos (FIG. 3, panels A-D). The 1262 25

nt mex-3 eDNA clone20 was divided into two segments, mex-3A and mex-3B with a short (325 nt) overlap. Similar results were obtained in experiments with no overlap between inhib­iting and probe segments. mex-3B antisense or dsRNA was injected into the gonads of adult animals, which were main- 30

tained under standard culture conditions for 24 hours before fixation and in situ hybridization (see Reference 5). The mex-3B dsRNA produced 100% embryonic arrest, while >90% of embryos from the antisense injections hatched. Antisense probes corresponding to mex-3A were used to assay distri- 35

bution of the endogenous mex-3 mRNA (dark stain). Four­cell stage embryos were assayed; similar results were observed from the 1 to 8 cell stage and in the germline of injected adults. The negative control (the absence of hybrid­ization probe) showed a lack of staining (FIG. 3A). Embryos 40

from uninjected parents showed a normal pattern of endog­enous mex-3 RNA (FIG. 3B). The observed pattern of mex-3 RNA was as previously described in Reference 20. Injection

22 FIGS. 5 A-C show a process and examples of genetic

inhibition following ingestion by C. elegans of dsRNAs from expressing bacteria. A general strategy for production of dsRNA is to clone segments of interest between flanking copies of the bacteriophage T7 promoter into a bacterial plasmid construct (FIG. SA). A bacterial strain (BL21/ DE3 ? 8 expressing the T7 polymerase gene from an inducible (Lac) promoter was used as a host. A nuclease-resistant dsRNA was detected in lysates oftransfected bacteria. Com­parable inhibition results were obtained with the two bacterial expression systems. A GFP-expressing C. elegans strain, PD4251 (see FIG. 2), was fed on a native bacterial host. These animals show a uniformly high level of GFP fluorescence in body muscles (FIG. SB). PD4251 animals were also reared on a diet of bacteria expressing dsRNA corresponding to the coding region for gfp. Under the conditions of this experi-ment, 12% of these animals showed dramatic decreases in GFP (FIG. SC). As an alternative strategy, single copies of the T7 promoter were used to drive expression of an inverted­duplication for a segment of the target gene, either unc-22 or gfp. This was comparably effective.

All references (e.g., books, articles, applications, and pat-ents) cited in this specification are indicative of the level of skill in the art and their disclosures are incorporated herein in their entirety. 1. Izant, J. & Weintraub, H. Cell36, 1007-1015 (1984). 2. Nellen, W. & Lichtenstein, C. TIES 18, 419-423 (1993). 3. Fire, A., eta!. Development 113, 503-514 (1991). 4. Guo, S. & Kemphues, K. CellS!, 611-620 (1995). 5. Seydoux, G. & Fire, A. Development 120, 2823-2834

(1994). 6. Ausubel, F., eta!. Current Protocols in Molecular Biology,

John Wiley N.Y. (1990). 7. Brenner, S. Genetics 77, 71-94 (1974). 8. Moerman, D. & Baillie, D. Genetics 91, 95-104 (1979). 9. Berrian, G., eta!. Genetics 134, 1097-1104 (1993). 10. Proud, C. TIES 20, 241-246 (1995). 11. Epstein H., eta!. J. Mol. Biol. 90 291-300 (1974). 12. Karn, J., eta!. Proc. Nat!. A cad. Sci. (U.S.A.) 80, 4253-

4257 (1983). 13. Doniach, T. & Hodgkin J. A. Dev. Biol. 106, 223-235

(1984). 14. Spence, A., eta!. Cell60, 981-990 (1990). 15. Krause, M., eta!. Cell63, 907-919 (1990). 16. Chen, L., eta!. Development, 120, 1631-1641 (1994). 17. Dibb, N.J., eta!. J. Mol. Biol. 205, 603-613 (1989). 18. Sulston, J ., et a!. Dev. Biol. 100, 64-119 (1983 ). 19. Sulston, J. & Horvitz, H. Dev. Biol. 82, 41-55 (1977).

of purified mex-3B antisense RNA produced at most a modest effect: the resulting embryos retained mex-3 mRNA, 45

although levels may have been somewhat less than wild type (FIG. 3C). In contrast, no mex-3 RNA was detected in embryos from parents injected with dsRNA corresponding to mex-3B (FIG. 3D). The scale of FIG. 3 is such that each embryo is approximately 50 f.tm in length.

Gene-specific inhibitory activity by unc-22A RNA was measured as a function of RNA structure and concentration (FIG. 4). Purified antisense and sense RNA from unc22A were injected individually or as an annealed mixture. "Con­trol" was an unrelated dsRNA (gfpG). Injected animals were 55

transferred to fresh culture plates 6 hours ( colunms labeled 1 ),

20. Draper B. W., eta!. Cell87, 205-216 (1996). 50 21. Sulston, J., eta!. Nature 356, 37-41 (1992).

15 hours (colunms labeled 2), 27 hours (colunms labeled 3), 41 hours ( colunms labeled 4 ), and 56 hours (columns labeled 5) after injection. Progeny grown to adulthood were scored for movement in their growth environment, then examined in 60

0.5 mM levamisole. The main graph indicates fractions in each behavioral class. Embryos in the uterus and already covered with an eggshell at the time of injection were not affected and, thus, are not included in the graph. The bottom­left diagram shows the genetically derived relationship 65

between unc-22 gene dosage and behavior based on analyses ofunc-22 heterozygotes and polyploids8

•3

.

22. Matzke, M. & Matzke, A. Plant Physiol. 107, 679-685 (1995).

23. Ratcliff, F., eta!. Science 276, 1558-1560 (1997). 24. Latham, K. Trends in Genetics 12,134-138 (1996). 25. Mello, C. & Fire, A. Methods in Cell Biology 48, 451-482

(1995). 26. Clark, D., eta!. Mol. Gen. Genet. 247, 367-378 (1995). 27. Chalfie, M., eta!. Science 263, 802-805 (1994). 28. Studier, F., et a!. Methods in Enzymology 185, 60-89

(1990). 29. Bowerman, B., eta!. Cell68, 1061-1075 (1992). 30. Mello, C. C., eta!. Cell70, 163-176 (1992). 31. van der Key!, H., eta!. Develop. Dynamics 201, 86-94

(1994). 32. Goedde!, D. V. Gene Expression Technology, Academic

Press, 1990.

Page 25: 112   andrew fire - 7622633 - genetic inhibition by double-stranded rna

US 7,622,633 B2 23

33. Kriegler, M. Gene Transfer and Expression, Stockton Press, 1990.

34. Murray, E. J. Gene Transfer and Expression Protocols, Humana Press, 1991.

TABLE 1

Effects of sense, antisense, and mixed RNAs on 12rogeny of injected animals.

Injected Gene and Segment Size RNA Fl Phenotype

unc-22 null mutants: strong unc-22 twitchers 7•

8

unc22Aa exon 21-22 742 sense wild type antisense wild type sense+ strong twitchers (100%) antisense

unc22B exon 27 1033 sense wild type antisense wild type sense+ strong twitchers (100%) antisense

unc22C exon 21-22b 785 sense+ strong twitchers (100%) antisense

fern-! fern-! null mutants: female (no sperm) 13

femlA exon lOc 531 sense hermaphrodite (98%) antisense hermaphrodite (>98%) sense+ female (72%) antisense

femlB intron 8 556 sense+ hermaphrodite (>98%) antisense

unc-54 unc-54 null mutants: paralyzed7,11

unc54A exon 6 576 sense wild type (100%) antisense wild type (100%) sense+ paralyzed (100%/ antisense

unc54B exon 6 651 sense wild type (100%) antisense wild type (100%) sense+ paralyzed (100%/ antisense

unc54C exon 1-5 1015 sense+ arrested embryos and larvae antisense (100%)

unc54D promoter 567 sense+ wild type (100%) antisense

unc54E intron 1 369 sense+ wild type (100%) antisense

unc54F intron 3 386 sense+ wild type (100%) antisense

hlh-1 null mutants: lumpy-dumpy hlh-1 larvae 16

hlhlA exons 1-6 1033 sense wild type ( <2% lpy-dpy) antisense wild type ( <2% lpy-dpy) sense+ lpy-dpy larvae (>90%)" antisense

hlhlB exons 1-2 438 sense+ lpy-dpy larvae (>80%)" antisense

hlhlC exons 4-6 299 sense+ lpy-dpy larvae (>80%)" antisense

hlhlD intron 1 697 sense+ wild type ( <2% lpy-dpy) antisense

myo-3 driven GFP transgenesf myo-3::NLS::gfp::lacZ makes nuclear GFP in body muscle

gfPG exons 2-5 730 sense nuclear GFP-LacZ pattern of parent strain

antisense nuclear GFP-LacZ pattern of parent strain

sense+ nuclear GFP-LacZ absent in antisense 98% of cells

lacZL exon 12-14 830 sense+ nuclear GFP-LacZ absent in antisense >95% of cells

10

15

20

25

30

35

40

45

50

55

60

65

24

TABLE !-continued

Effects of sense, antisense, and mixed RNAs on progeny of injected animals.

Injected Gene and Segment Size RNA Fl Phenotype

makes mitochondrial GFP in body myo-3: :MtLS: :gfP muscle

gfpG exons 2-5 730 sense mitochondrial GFP pattern of parent strain

antisense mitochondrial GFP pattern of parent strain

sense+ mitochondrial GFP absent antisense in 98% of cells

lacZL exon 12-14 830 sense+ mitochondrial GFP pattern antisense of parent strain

Legend of Table 1 Each RNA was injected into 6-10 adult hermaphrodites (0.5-1 x 106 mol-ecules into each gonad arm). After 4-6 hours (to clear pre-fertilized eggs from the uterus) injected animals were transferred and eggs collected for 20-22 hours. Progeny phenotypes were scored upon hatching and subse-quently at 12-24 hour intervals. aTo obtain a semi-quantitative assessment of the relationship between RNA dose and phenotypic response, we injected each unc22A RNA preparation at a series of different concentrations. At the highest dose tested (3.6 x 106

molecules per gonad), the individual sense and antisense unc22A prepara-lions produced some visible twitching (1% and 11% of progeny respec-lively). Comparable doses of ds-unc22A RNA produced visible twitching in all progeny, while a 120-fold lower dose of ds-unc22A RNA produced vis-ible twitching in 30% of progeny. bunc22C also carries the intervening intron (43 nt). cfemlA also carries a portion (131 nt) of intron 10. d Animals in the first affected broods (laid at 4-24 hours after injection) showed movement defects indistinguishable from those of null mutants in unc-54. A variable fraction of these animals (25-75%) failed to lay eggs (an-other phenotype ofunc-54 null mutants), while the remainder of the para-lyzed animals were egg-laying positive. This may indicate partial inhibition ofunc-54 activity in vulval muscles. Animals from later broods frequently exhibit a distinct partial loss-of-function phenotype, with contractility in a subset of body wall muscles. ePhenotypes ofhlh-1 inhibitory RNA include arrested embryos and partially elongated Lllarvae (the hlh-1 null phenotype) seen in virtually all progeny from injection of ds-hlhlA and about half of the affected animals from ds-hlhlB and ds-hlhlC) and a set ofless severe defects (seen with the remainder of the animals from ds-hlhlB and ds-hlhlC). The less severe phe-notypes are characteristic of partial loss of function for hlh-1. -'The host for these injections, PD4251, expresses both mitochondrial GFP and nuclear GFP-LacZ. This allows simultaneous assay for inhibition of gfp (loss of all fluorescence) and lacA (loss of nuclear fluorescence). The table describes scoring of animals as L1 larvae. ds-gfpG caused a loss of GFP in all but 0-3 of the 85 body muscles in these larvae. As these animals mature to adults, GFP activity was seen in 0-5 additional bodywall muscles in the eight vulval muscles.

TABLE2

Effect of injection point on genetic inhibition in injected animals and their 12rogeny.

Site of Injected animal dsRNA injection phenotype Progeny Phenotype

None gonad or body no twitching no twitching cavity

None gonad or body strong nuclear & strong nuclear & cavity mitochondrial mitochondrial

GFP GFP unc22B Gonad weak twitchers strong twitchers unc22B Body Cavity weak twitchers strong twitchers

Head unc22B Body Cavity weak twitchers strong twitchers

Tail gfpG Gonad lower nuclear & rare or absent nuclear &

mitochondrial mitochondrial GFP GFP

Page 26: 112   andrew fire - 7622633 - genetic inhibition by double-stranded rna

US 7,622,633 B2

dsRNA

gfPG

lacZL

lacZL

25

TABLE 2-continued

Effect of injection point on genetic inhibition in injected animals and their progeny.

Site of Injected animal injection phenotype Progeny Phenotype

Body Cavity lower nuclear & rare or absent nuclear & Tail mitochondrial mitochondrial GFP

GFP Gonad lower nuclear GFP rare or absent nuclear

GFP Body Cavity lower nuclear GFP rare or absent nuclear Tail GFP

TABLE3

C. elegans can respond in a gene-specific manner to environmental dsRNA.

Germline Bacterial Food Movement Phenotype GFP-Transgene Expression

BL21(DE3) 0% twitch <1% female <1% faint GFP BL21(DE3) 0% twitch 43% female <1% faint GFP [fern-! dsRNA] BL21(DE3) 85% twitch <1% female <1% faint GFP [unc22 dsRNA] BL21(DE3) 0% twitch <1% female 12% faint GFP [gfp dsRNA]

dsRNA

unc-22 pos-1 sqt-3

TABLE4

Effects of bathing C. elegans in a solution containing dsRNA.

Biological Effect

Twitching (similar to partial loss ofunc-22 function) Embryonic arrest (similar to loss of pos-1 function) Shortened body (Dpy) (similar to partial loss of sqt-3 function)

In Table 2, gonad injections were carried out into the GFP reporter strain PD4251, which expresses both mitochondrial GFP and nuclear GFP-LacZ. This allowed simultaneous assay of inhibition with gfp (fainter overall fluorescence), lacZ (loss of nuclear fluorescence), and unc-22 (twitching). Body cavity injections were carried out into the tail region, to minimize accidental injection of the gonad; equivalent results have been observed with injections into the anterior region of the body cavity. An equivalent set of injections was also performed into a single gonad arm. For all sites of injection, the entire progeny brood showed phenotypes identical to

10

26 those described in Table 1. This included progeny produced from both injected and uninjected gonad arms. Injected ani­mals were scored three days after recovery and showed some­what less dramatic phenotypes than their progeny. This could in part be due to the persistence of products already present in the injected adult. After ds-unc22B injection, a fraction of the injected animals twitch weakly under standard growth con­ditions (10 out of21 animals). Levamisole treatment led to twitching of 100% (21/21) of these animals. Similar effects were seen with ds-unc22A. Injections of ds-gfpG or ds-lacZL produced a dramatic decrease (but not elimination) of the corresponding GFP reporters. In some cases, isolated cells or parts of animals retained strong GFP activity. These were most frequently seen in the anterior region and around the

15 vulva. Injections of ds-gfpG and ds-lacZL produced no twitching, while injections of ds-unc22A produced no change in GFP fluorescence pattern.

While the present invention has been described in connec­tion with what is presently considered to be practical and

20 preferred embodiments, it is understood that the invention is not to be limited or restricted to the disclosed embodiments but, on the contrary, is intended to cover various modifica­tions and equivalent arrangements included within the spirit and scope of the appended claims.

25 Thus it is to be understood that variations in the described invention will be obvious to those skilled in the art without departing from the novel aspects of the present invention and such variations are intended to come within the scope of the present invention.

30 We claim: 1. A method to inhibit expression of a target gene in a cell

in a plant comprising contacting the cell with an expression construct that produces at least two separate ribonucleic acids (RNAs) in an amount sufficient to inhibit the expression of a

35 target gene, wherein the RNAs form a double-stranded struc­ture containing separate complementary strands, wherein the first strand corresponds to a nucleotide sequence of the target gene and the second ribonucleotide strand is complementary to the nucleotide sequence of the target gene, wherein the first

40 and the second ribonucleotide strands are separate comple­mentary sequences that hybridize to each other to form said double-stranded structure, which inhibits expression of the target gene.

2. The method of claim 1, wherein said plant is selected 45 from the group consisting of monocots, dicots and gynmo­

sperms. 3. The method of claim 1, wherein said plant is selected

from the group consisting of arabidopsis, corn, wheat, tobacco, soybean, cauliflower, potato, pine, rubber, and oak.

* * * * *

Page 27: 112   andrew fire - 7622633 - genetic inhibition by double-stranded rna

UNITED STATES PATENT AND TRADEMARK OFFICE

CERTIFICATE OF CORRECTION

PATENT NO. : 7,622,633 B2 Page 1 of 1 APPLICATION NO. : 10/283267 DATED : November 24, 2009 INVENTOR(S) : Fire et al.

It is certified that error appears in the above-identified patent and that said Letters Patent is hereby corrected as shown below:

On the Title Page:

The first or sole Notice should read --

Subjectto any disclaimer, the term of this patent is extended or adjusted under 35 U.S.C. 154(b) by 1953 days.

Signed and Sealed this

Fourteenth Day of December, 2010

David J. Kappas Director of the United States Patent and Trademark Office